Review Article

A Comprehensive Review on Liver Targeting: Emphasis on Nanotechnology- based Molecular Targets and Receptors Mediated Approaches

Author(s): Ashwini Kumar Mishra, Mukesh Pandey, Hitesh Kumar Dewangan, Neha SL and Pravat Kumar Sahoo*

Volume 23, Issue 15, 2022

Published on: 30 September, 2022

Page: [1381 - 1405] Pages: 25

DOI: 10.2174/1389450123666220906091432

Price: $65

Abstract

Background: The pathogenesis of hepatic diseases involves several cells, which complicates the delivery of pharmaceutical agents. Many severe liver diseases affecting the worldwide population cannot be effectively treated. Major hindrances or challenges are natural physiological barriers and non-specific targeting of drugs administered, leading to inefficient treatment. Hence, there is an earnest need to look for novel therapeutic strategies to overcome these hindrances. A kind of literature has reported that drug safety and efficacy are incredibly raised when a drug is incorporated inside or attached to a polymeric material of either hydrophilic or lipophilic nature. This has driven the dynamic investigation for developing novel biodegradable materials, drug delivery carriers, target-specific drug delivery systems, and many other novel approaches.

Objective: Present review is devoted to summarizing receptor-based liver cell targeting using different modified novel synthetic drug delivery carriers. It also highlights recent progress in drug targeting to diseased liver mediated by various receptors, including asialoglycoprotein, mannose and galactose receptor, Fc receptor, low-density lipoprotein, glycyrrhetinic, and bile acid receptor. The essential consideration is given to treating liver cancer targeting using nanoparticulate systems, proteins, viral and non-viral vectors, homing peptides and gene delivery.

Conclusion: Receptors based targeting approach is one such approach that was explored by researchers to develop novel formulations which can ensure site-specific drug delivery. Several receptors are on the surfaces of liver cells, which are highly overexpressed in various disease conditions. They all are helpful for the treatment of liver cancer.

Keywords: Hepatic diseases, liver, cancer, receptors, targeting, nanoparticulate systems.

Graphical Abstract
[1]
Mokdad AA, Lopez AD, Shahraz S, et al. Liver cirrhosis mortality in 187 countries between 1980 and 2010: A systematic analysis. BMC Med 2014; 12(1): 145.
[http://dx.doi.org/10.1186/s12916-014-0145-y] [PMID: 25242656]
[2]
Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol 2019; 70(1): 151-71.
[http://dx.doi.org/10.1016/j.jhep.2018.09.014] [PMID: 30266282]
[3]
Massey VL, Arteel GE. Acute alcohol induced liver injury. Front Physiol 2012; 3: 193.
[http://dx.doi.org/10.3389/fphys.2012.00193] [PMID: 22701432]
[4]
Dey A, Cederbaum AI. Alcohol and oxidative liver injury. Hepatology 2006; 43(S1): 63-74.
[http://dx.doi.org/10.1002/hep.20957] [PMID: 16447273]
[5]
Chen X. Protective effects of quercetin on liver injury induced by ethanol. Pharmacogn Mag 2010; 6(22): 135-41.
[http://dx.doi.org/10.4103/0973-1296.62900] [PMID: 20668581]
[6]
George A. Global status report on alcohol 2004. Can J Public Health 2006; 97(5): 408.
[7]
Sacco R, Eggenhoffner R, Giacomelli L. Glutathione in the treatment of liver diseases: Insights from clinical practice. Minerva Gastroenterol Dietol 2016; 62(4): 316-24.
[PMID: 27603810]
[8]
Dewangan HK, Sharma A, Mishra A, Singour P. Mucoadhesive microspheres of atorvastatin calcium: Rational design, evaluation and enhancement of bioavailability. Indian J Pharm Educ Res 2021; 55(3): S733-41.
[http://dx.doi.org/10.5530/ijper.55.3s.180]
[9]
Marchesini G, Roden M, Vettor R. Response to: Comment to “EASL-EASD-EASO Clinical Practice Guidelines for the management of non alcoholic fatty liver disease”. J Hepatol 2017; 66(2): 466-7.
[http://dx.doi.org/10.1016/j.jhep.2016.11.002] [PMID: 27856217]
[10]
Peverill W, Powell L, Skoien R. Evolving concepts in the pathogenesis of NASH: Beyond steatosis and inflammation. Int J Mol Sci 2014; 15(5): 8591-638.
[http://dx.doi.org/10.3390/ijms15058591] [PMID: 24830559]
[11]
Marzuillo P, Grandone A, Perrone L, Miraglia Del Giudice E. Understanding the pathophysiological mechanisms in the pediatric non alcoholic fatty liver disease: The role of genetics. World J Hepatol 2015; 7(11): 1439-43.
[http://dx.doi.org/10.4254/wjh.v7.i11.1439] [PMID: 26085904]
[12]
Dewangan HK, Singh S, Maurya L, Srivastava A. Hepatitis B antigen loaded biodegradable polymeric nanoparticles: Formulation optimization and in vivo immunization in BALB/c Mice. Curr Drug Deliv 2018; 15(8): 1204-15.
[http://dx.doi.org/10.2174/1567201815666180604110457] [PMID: 29866006]
[13]
Margaret C. Global Hepatitis Report 2017. World Health Organization 2017. Licence: CC BY-NC-SA 30 IGO.
[14]
Dewangan HK, Pandey T, Singh S. Nanovaccine for immunotherapy and reduced hepatitis-B virus in humanized model. Artif Cells Nanomed Biotechnol 2018; 46(8): 2033-42.
[PMID: 29179600]
[15]
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394-424.
[http://dx.doi.org/10.3322/caac.21492] [PMID: 30207593]
[16]
Dewangan HK. Rational application of nanoadjuvant for mucosal vaccine delivery system. J Immunol Methods 2020; 481-482: 112791.
[http://dx.doi.org/10.1016/j.jim.2020.112791] [PMID: 32387695]
[17]
Dewangan HK. Albumin as natural versatile drug carrier for various diseases treatment. Sustain Agric Res 2020; 43: 239-68.
[18]
Garg A, Dewangan HK. Nanoparticles as adjuvants in vaccine delivery. Crit Rev Ther Drug Carrier Syst 2020; 37(2): 183-204.
[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.2020033273]
[19]
Mishra N, Yadav NP, Rai VK, et al. Efficient hepatic delivery of drugs: Novel strategies and their significance. BioMed Res Int 2013; 2013: 1-20.
[http://dx.doi.org/10.1155/2013/382184] [PMID: 24286077]
[20]
Sultatos L. First Pass Effect InxPharm: The Comprehensive Pharmacology Reference. Elsevier Inc. 2007; pp. 1-2.
[http://dx.doi.org/10.1016/B978-008055232-3.60022-4]
[21]
Thakkar N, Slizgi JR, Brouwer KLR. Effect of liver disease on hepatic transporter expression and function. J Pharm Sci 2017; 106(9): 2282-94.
[http://dx.doi.org/10.1016/j.xphs.2017.04.053] [PMID: 28465155]
[22]
Poelstra K, Prakash J, Beljaars L. Drug targeting to the diseased liver. J Cont Release 2012; 161(2): 188-97.
[http://dx.doi.org/10.1016/j.jconrel.2012.02.011] [PMID: 22370583]
[23]
Lewis D. Human cytochromes P450 associated with the phase 1 metabolism of drugs and other xenobiotics: A compilation of substrates and inhibitors of the CYP1, CYP2 and CYP3 families. Curr Med Chem 2003; 10(19): 1955-72.
[http://dx.doi.org/10.2174/0929867033456855] [PMID: 12871098]
[24]
Montfoort J, Hagenbuch B, Groothuis G, Koepsell H, Meier P, Meijer D. Drug uptake systems in liver and kidney. Curr Drug Metab 2003; 4(3): 185-211.
[http://dx.doi.org/10.2174/1389200033489460] [PMID: 12769665]
[25]
Erion MD. Prodrugs for liver targeted drug delivery. In: Stella VJ, Borchardt RT, Hageman MJ, Oliyai R, Maag H, Tilley JW, Eds. Prodrugs biotechnology: Pharmaceutical aspects. New York: Springer 2007; p. 3.
[http://dx.doi.org/10.1007/978-0-387-49785-3_16]
[26]
Stella V, Borchardt R, Hageman M, Oliyai R, Maag H, Tilley J. Prodrugs: Challenges and rewards. Springer Science Business Media 2007.
[27]
Boddy A, Aarons L, Petrak K. Efficiency of drug targeting: Steady state considerations using a three compartment model. Pharm Res 1989; 6(5): 367-72.
[http://dx.doi.org/10.1023/A:1015971113161] [PMID: 2748526]
[28]
Hashida M, Opanasopit P, Nishikawa M. Factors affecting drug and gene delivery: Effects of interaction with blood components. Crit Rev Ther Drug Carrier Syst 2002; 19(3): 191-234.
[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v19.i3.10] [PMID: 12627613]
[29]
Morgan PJ, Harding SE, Petrak K. Interactions of a model block copolymer drug delivery system with two serum proteins and myoglobin. Biochem Soc Trans 1990; 18(5): 1021-2.
[http://dx.doi.org/10.1042/bst0181021] [PMID: 2083644]
[30]
Ivanenkov YA, Maklakova SY, Beloglazkina EK, et al. Development of liver cell targeted drug delivery systems: Experimental approaches. Russ Chem Rev 2017; 86(8): 750-76.
[http://dx.doi.org/10.1070/RCR4707]
[31]
Wu F, Li X, Jiang B, et al. Glycyrrhetinic acid functionalized nanoparticles for drug delivery to liver cancer. J Biomed Nanotechnol 2018; 14(11): 1837-52.
[http://dx.doi.org/10.1166/jbn.2018.2638] [PMID: 30165922]
[32]
Lakshmi Singh S, Vijayakumar MR, Dewangan HK. Lipid based Aqueous Core Nanocapsules (ACNs) for encapsulating hydrophilic vinorelbine bitartrate: Preparation, optimization, characterization and in vitro safety assessment for intravenous administration. Curr Drug Deliv 2018; 15(9): 1284-93.
[http://dx.doi.org/10.2174/1567201815666180716112457] [PMID: 30009708]
[33]
Zhang J, Zhang M, Ji J, et al. Glycyrrhetinic acid mediated polymeric drug delivery targeting the acidic microenvironment of hepatocellular carcinoma. Pharm Res 2015; 32(10): 3376-90.
[http://dx.doi.org/10.1007/s11095-015-1714-2] [PMID: 26148773]
[34]
Wu J, Nantz MH, Zern MA. Targeting hepatocytes for drug and gene delivery emerging novel approaches and applications. Front Biosci 2002; 7(4): A806.
[http://dx.doi.org/10.2741/A806] [PMID: 11861224]
[35]
Huang X, Leroux JC, Castagner B. Well defined multivalent ligands for hepatocytes targeting via asialoglycoprotein receptor. Bioconjug Chem 2017; 28(2): 283-95.
[http://dx.doi.org/10.1021/acs.bioconjchem.6b00651] [PMID: 27966887]
[36]
Liang HF, Chen CT, Chen SC, et al. Paclitaxel loaded poly(γ-glutamic acid)-poly(lactide) nanoparticles as a targeted drug delivery system for the treatment of liver cancer. Biomaterials 2006; 27(9): 2051-9.
[http://dx.doi.org/10.1016/j.biomaterials.2005.10.027] [PMID: 16307794]
[37]
Gupta PN, Mahor S, Rawat A, Khatri K, Goyal A, Vyas SP. Lectin anchored stabilized biodegradable nanoparticles for oral immunization. Int J Pharm 2006; 318(1-2): 163-73.
[http://dx.doi.org/10.1016/j.ijpharm.2006.03.017] [PMID: 16621367]
[38]
Bibby DC, Talmadge JE, Dalal MK, et al. Pharmacokinetics and biodistribution of RGD-targeted doxorubicin loaded nanoparticles in tumor-bearing mice. Int J Pharm 2005; 293(1-2): 281-90.
[http://dx.doi.org/10.1016/j.ijpharm.2004.12.021] [PMID: 15778066]
[39]
Stella B, Arpicco S, Peracchia MT, et al. Design of folic acid conjugated nanoparticles for drug targeting. J Pharm Sci 2000; 89(11): 1452-64.
[http://dx.doi.org/10.1002/1520-6017(200011)89:11<1452:AID-JPS8>3.0.CO;2-P] [PMID: 11015690]
[40]
Grobmyer SR, Iwakuma N, Sharma P, Moudgil BM. What is cancer nanotechnology? Methods Mol Biol 2010; 624: 1-9.
[http://dx.doi.org/10.1007/978-1-60761-609-2_1] [PMID: 20217585]
[41]
Iden DL, Allen TM. In vitro and in vivo comparison of immunoliposomes made by conventional coupling techniques with those made by a new post insertion approach. Biochim Biophys Acta Biomembr 2001; 1513(2): 207-16.
[http://dx.doi.org/10.1016/S0005-2736(01)00357-1] [PMID: 11470092]
[42]
Pathak K, Vaidya A. Avidin based nanoparticles for drug delivery.Applications of Nanocomposite Materials in Drug Delivery. Woodhead Publishing 2018; pp. 163-90.
[http://dx.doi.org/10.1016/B978-0-12-813741-3.00007-8]
[43]
Zeng X, Sun YX, Zhang XZ, Zhuo RX. Biotinylated disulfide containing PEI/avidin bioconjugate shows specific enhanced transfection efficiency in HepG2 cells. Org Biomol Chem 2009; 7(20): 4201-10.
[http://dx.doi.org/10.1039/b910831a] [PMID: 19795058]
[44]
Damont A, Boisgard R, Dollé F, Hollocou M, Kuhnast B. Avidin/biotin bioinspired platform for dual in vivo F-PET/NIRF molecular imaging. Bioconjug Chem 2017; 28(10): 2524-9.
[http://dx.doi.org/10.1021/acs.bioconjchem.7b00536] [PMID: 28931274]
[45]
Violatto MB, Casarin E, Talamini L, et al. Dexamethasone conjugation to biodegradable avidin nucleic acid nano assemblies promotes selective liver targeting and improves therapeutic efficacy in an autoimmune hepatitis murine model. ACS Nano 2019; 13(4): 4410-23.
[http://dx.doi.org/10.1021/acsnano.8b09655] [PMID: 30883091]
[46]
Yu F, Jiang T, Zhang J, Cheng L, Wang S. Galactosylated liposomes as oligodeoxynucleotides carrier for hepatocyte selective targeting. Pharmazie 2007; 62(7): 528-33.
[PMID: 17718195]
[47]
Wu J, Liu P, Zhu JL, Maddukuri S, Zern MA. Increased liver uptake of liposomes and improved targeting efficacy by labeling with asialofetuin in rodents. Hepatology 1998; 27(3): 772-8.
[http://dx.doi.org/10.1002/hep.510270319] [PMID: 9500706]
[48]
Opanasopit P, Sakai M, Nishikawa M, Kawakami S, Yamashita F, Hashida M. Inhibition of liver metastasis by targeting of immunomodulators using mannosylated liposome carriers. J Control Release 2002; 80(1-3): 283-94.
[http://dx.doi.org/10.1016/S0168-3659(02)00006-8] [PMID: 11943405]
[49]
Zhang XQ, Wang XL, Zhang PC, et al. Galactosylated ternary DNA/polyphosphoramidate nanoparticles mediate high gene transfection efficiency in hepatocytes. J Control Release 2005; 102(3): 749-63.
[http://dx.doi.org/10.1016/j.jconrel.2004.10.024] [PMID: 15681095]
[50]
Kang JH, Oishi J, Kim JH, et al. Hepatoma targeted gene delivery using a tumor cell specific gene regulation system combined with a human liver cell specific bionanocapsule. Nanomedicine 2010; 6(4): 583-9.
[http://dx.doi.org/10.1016/j.nano.2010.01.007] [PMID: 20138242]
[51]
Arima H, Yamashita S, Mori Y, et al. In vitro and in vivo gene delivery mediated by Lactosylated dendrimer/alpha cyclodextrin conjugates (G2) into hepatocytes. J Control Release 2010; 146(1): 106-17.
[http://dx.doi.org/10.1016/j.jconrel.2010.05.030] [PMID: 20678990]
[52]
Kaneo Y, Tanaka T, Nakano T, Yamaguchi Y. Evidence for receptor mediated hepatic uptake of pullulan in rats. J Control Release 2001; 70(3): 365-73.
[http://dx.doi.org/10.1016/S0168-3659(00)00368-0] [PMID: 11182206]
[53]
Tolleshaug H, Berg T, Blomhoff R. Uptake of mannose terminated glycoproteins in isolated rat liver cells. Evidence for receptor mediated endocytosis in hepatocytes. Biochem J 1984; 223(1): 151-60.
[http://dx.doi.org/10.1042/bj2230151] [PMID: 6497838]
[54]
Fielding CJ. Lipoprotein receptors, plasma cholesterol metabolism, and the regulation of cellular free cholesterol concentration. FASEB J 1992; 6(13): 3162-8.
[http://dx.doi.org/10.1096/fasebj.6.13.1327930] [PMID: 1327930]
[55]
Dalen DPPV, De Leeuw AM, Brouwer A, Knook DL. Rat liver endothelial cells have a greater capacity than kupffer cells to endocytoseN-acetylglucosamine- and mannose-terminated glycoproteins. Hepatology 1987; 7(4): 672-9.
[http://dx.doi.org/10.1002/hep.1840070410] [PMID: 3301616]
[56]
Fadden AJ, Holt OJ, Drickamer K. Molecular characterization of the rat Kupffer cell glycoprotein receptor. Glycobiology 2003; 13(7): 529-37.
[http://dx.doi.org/10.1093/glycob/cwg068] [PMID: 12672702]
[57]
Lehrman MA, Hill RL. The binding of fucose containing glycoproteins by hepatic lectins. Purification of a fucose-binding lectin from rat liver. J Biol Chem 1986; 261(16): 7419-25.
[http://dx.doi.org/10.1016/S0021-9258(17)38408-9] [PMID: 3711093]
[58]
Fridman WH. Fc receptors and immunoglobulin binding factors. FASEB J 1991; 5(12): 2684-90.
[http://dx.doi.org/10.1096/fasebj.5.12.1916092] [PMID: 1916092]
[59]
Helmy KY, Katschke KJ Jr, Gorgani NN, et al. CRIg: A macrophage complement receptor required for phagocytosis of circulating pathogens. Cell 2006; 124(5): 915-27.
[http://dx.doi.org/10.1016/j.cell.2005.12.039] [PMID: 16530040]
[60]
Van Berkel TJ, De Rijke YB, Kruijt JK. Different fate in vivo of oxidatively modified low density lipoprotein and acetylated low density lipoprotein in rats. Recognition by various scavenger receptors on Kupffer and endothelial liver cells. J Biol Chem 1991; 266(4): 2282-9.
[http://dx.doi.org/10.1016/S0021-9258(18)52241-9] [PMID: 1989982]
[61]
Leitinger B, Hohenester E. Mammalian collagen receptors. Matrix Biol 2007; 26(3): 146-55.
[http://dx.doi.org/10.1016/j.matbio.2006.10.007] [PMID: 17141492]
[62]
Weigel JA, Weigel PH. Characterization of the recombinant rat 175-kDa hyaluronan receptor for endocytosis (HARE). J Biol Chem 2003; 278(44): 42802-11.
[http://dx.doi.org/10.1074/jbc.M307201200] [PMID: 12933790]
[63]
Li FQ, Su H, Chen X, et al. Mannose 6-phosphate modified bovine serum albumin nanoparticles for controlled and targeted delivery of sodium ferulate for treatment of hepatic fibrosis. J Pharm Pharmacol 2010; 61(9): 1155-61.
[http://dx.doi.org/10.1211/jpp.61.09.0004] [PMID: 19703364]
[64]
Bansal R, Prakash J, Ruijter M, Beljaars L, Poelstra K. Peptide modified albumin carrier explored as a novel strategy for a cell-specific delivery of interferon gamma to treat liver fibrosis. Mol Pharm 2011; 8(5): 1899-909.
[http://dx.doi.org/10.1021/mp200263q] [PMID: 21800888]
[65]
Stockert RJ. The asialoglycoprotein receptor: Relationships between structure, function, and expression. Physiol Rev 1995; 75(3): 591-609.
[http://dx.doi.org/10.1152/physrev.1995.75.3.591] [PMID: 7624395]
[66]
Prakash TP, Graham MJ, Yu J, et al. Targeted delivery of antisense oligonucleotides to hepatocytes using triantennary N -acetyl galactosamine improves potency 10-fold in mice. Nucleic Acids Res 2014; 42(13): 8796-807.
[http://dx.doi.org/10.1093/nar/gku531] [PMID: 24992960]
[67]
Kim Y, Jo M, Schmidt J, et al. Enhanced potency of GalNAc-conjugated antisense oligonucleotides in hepatocellular cancer models. Mol Ther 2019; 27(9): 1547-57.
[http://dx.doi.org/10.1016/j.ymthe.2019.06.009] [PMID: 31303442]
[68]
D’Erasmo L, Gallo A, Di Costanzo A, Bruckert E, Arca M. Evaluation of efficacy and safety of antisense inhibition of apolipoprotein C-III with volanesorsen in patients with severe hypertriglyceridemia. Expert Opin Pharmacother 2020; 21(14): 1675-84.
[http://dx.doi.org/10.1080/14656566.2020.1787380] [PMID: 32646313]
[69]
Lee RJ, Lee RJ, Zhang , et al. Lactosylated liposomes for targeted delivery of doxorubicin to hepatocellular carcinoma. Int J Nanomedicine 2012; 7: 5465-74.
[http://dx.doi.org/10.2147/IJN.S33965] [PMID: 23093902]
[70]
Seymour LW, Ferry DR, Anderson D, et al. Hepatic drug targeting: Phase I evaluation of polymer-bound doxorubicin. J Clin Oncol 2002; 20(6): 1668-76.
[http://dx.doi.org/10.1200/JCO.2002.20.6.1668] [PMID: 11896118]
[71]
Dewangan HK, Singh S, Mishra R, Dubey RK. A review on application of nanoadjuvant as delivery system. Int J Appl Pharm 2020; 12(4): 24-33.
[http://dx.doi.org/10.22159/ijap.2020v12i4.36856]
[72]
Dewangan HK, Tomar S. Nanovaccine for transdermal delivery system. J Drug Deliv Sci Technol 2022; 67: 102988.
[http://dx.doi.org/10.1016/j.jddst.2021.102988]
[73]
Zhang Y, Wu X, Hou C, et al. Dual responsive dithio polydopamine coated porous CeO2 nanorods for targeted and synergistic drug delivery. Int J Nanomedicine 2018; 13: 2161-73.
[http://dx.doi.org/10.2147/IJN.S152002] [PMID: 29695903]
[74]
Ivanenkov YA, Majouga AG, Petrov RA, et al. Synthesis and biological evaluation of novel doxorubicin-containing ASGP-R-targeted drug-conjugates. Bioorg Med Chem Lett 2018; 28(3): 503-8.
[http://dx.doi.org/10.1016/j.bmcl.2017.12.004] [PMID: 29254645]
[75]
Tamura R, Uemoto S, Tabata Y. Augmented liver targeting of exosomes by surface modification with cationized pullulan. Acta Biomater 2017; 57: 274-84.
[http://dx.doi.org/10.1016/j.actbio.2017.05.013] [PMID: 28483695]
[76]
Pathak PO, Nagarsenker MS, Barhate CR, et al. Cholesterol anchored arabinogalactan for asialoglycoprotein receptor targeting: synthesis, characterization, and proof of concept of hepatospecific delivery. Carbohydr Res 2015; 408: 33-43.
[http://dx.doi.org/10.1016/j.carres.2015.03.003] [PMID: 25841057]
[77]
Zhong Y, Yang W, Sun H, et al. Ligand directed reduction sensitive shell sheddable biodegradable micelles actively deliver doxorubicin into the nuclei of target cancer cells. Biomacromolecules 2013; 14(10): 3723-30.
[http://dx.doi.org/10.1021/bm401098w] [PMID: 23998942]
[78]
Dhande YK, Wagh BS, Hall BC, Sprouse D, Hackett PB, Reineke TM. N -acetylgalactosamine block copolycations form stable polyplexes with plasmids and promote liver targeted delivery. Biomacromolecules 2016; 17(3): 830-40.
[http://dx.doi.org/10.1021/acs.biomac.5b01555] [PMID: 26854615]
[79]
Biessen EL, Noorman F, Teijlingen ME, et al. Lysine based cluster mannosides that inhibit ligand binding to the human mannose receptor at nanomolar concentration. J Biol Chem 1996; 271(45): 28024-30.
[http://dx.doi.org/10.1074/jbc.271.45.28024] [PMID: 8910412]
[80]
Ganbold T, Baigude H. Design of mannose-functionalized curdlan nanoparticles for macrophage targeted siRNA delivery. ACS Appl Mater Interfaces 2018; 10(17): 14463-74.
[http://dx.doi.org/10.1021/acsami.8b02073] [PMID: 29648784]
[81]
Arteta B, Lasuen N, Lopategi A, Sveinbjörnsson B, Smedsrød B, Vidal VF. Colon carcinoma cell interaction with liver sinusoidal endothelium inhibits organ-specific antitumor immunity through interleukin-1-induced mannose receptor in mice. Hepatology 2010; 51(6): 2172-82.
[http://dx.doi.org/10.1002/hep.23590] [PMID: 20513002]
[82]
Zhang J, Song H, Ji S, et al. NO prodrug-conjugated, self assembled, pH responsive and galactose receptor targeted nanoparticles for co-delivery of nitric oxide and doxorubicin. Nanoscale 2018; 10(9): 4179-88.
[http://dx.doi.org/10.1039/C7NR08176F] [PMID: 29442103]
[83]
Longmuir KJ, Robertson RT, Haynes SM, Baratta JL, Waring AJ. Effective targeting of liposomes to liver and hepatocytes in vivo by incorporation of a Plasmodium amino acid sequence. Pharm Res 2006; 23(4): 759-69.
[http://dx.doi.org/10.1007/s11095-006-9609-x] [PMID: 16550476]
[84]
Ma K, Shen H, Shen S, et al. Development of a successive targeting liposome with multi-ligand for efficient targeting gene delivery. J Gene Med 2011; 13(5): 290-301.
[http://dx.doi.org/10.1002/jgm.1569] [PMID: 21574214]
[85]
Wu F, Wuensch SA, Azadniv M, Ebrahimkhani MR, Crispe IN. Galactosylated LDL nanoparticles: A novel targeting delivery system to deliver antigen to macrophages and enhance antigen specific T cell responses. Mol Pharm 2009; 6(5): 1506-17.
[http://dx.doi.org/10.1021/mp900081y] [PMID: 19637876]
[86]
Zhang F, Wu Q, Chen ZC, Zhang M, Lin XF. Hepatic targeting microcapsules construction by self assembly of bioactive galactose branched polyelectrolyte for controlled drug release system. J Colloid Interface Sci 2008; 317(2): 477-84.
[http://dx.doi.org/10.1016/j.jcis.2007.09.065] [PMID: 17931643]
[87]
Yadav D, Dewangan HK. Pegylation: An important approach for novel drug delivery system. J Biomater Sci Polym Ed 2020; 3: 1-15.
[PMID: 32942961]
[88]
Nie H, Qiu B, Yang QX, et al. Effect of gal/GalNAc regioisomerism in galactosylated liposomes on asialoglycoprotein receptor mediated hepatocyte selective targeting in vivo. J Liposome Res 2021; 31(1): 79-89.
[http://dx.doi.org/10.1080/08982104.2019.1682606] [PMID: 31691619]
[89]
Shen H, Zhang M, Kaita K, Minuk GY, Rempel J, Gong Y. Expression of Fc fragment receptors of immunoglobulin G (Fc?Rs) in rat hepatic stellate cells. Dig Dis Sci 2005; 50(1): 181-7.
[http://dx.doi.org/10.1007/s10620-005-1298-5] [PMID: 27837389]
[90]
Wang Y, Tian Z, Thirumalai D, Zhang X. Neonatal Fc receptor (FcRn): A novel target for therapeutic antibodies and antibody engineering. J Drug Target 2014; 22(4): 269-78.
[http://dx.doi.org/10.3109/1061186X.2013.875030] [PMID: 24404896]
[91]
Yu D, Leja JJ, Loskog A, et al. Preclinical evaluation of advince, an oncolytic adenovirus adapted for treatment of liver metastases from neuroendocrine cancer. Neuroendocrinology 2017; 105(1): 54-66.
[http://dx.doi.org/10.1159/000448430] [PMID: 27442441]
[92]
Jeon H, Blacklow SC. Structure and physiologic function of the low-density lipoprotein receptor. Annu Rev Biochem 2005; 74(1): 535-62.
[http://dx.doi.org/10.1146/annurev.biochem.74.082803.133354] [PMID: 15952897]
[93]
Zhang XN, Zhang C, Zhu Q, et al. N-Succinyl chitosan nanoparticles coupled with low-density lipoprotein for targeted osthole loaded delivery to low density lipoprotein receptor-rich tumors. Int J Nanomedicine 2014; 9: 2919-32.
[http://dx.doi.org/10.2147/IJN.S59799] [PMID: 24966673]
[94]
Kiso Y, Tohkin M, Hikino H. Mechanism of antihepatotoxic activity of atractylon, I: Effect on free radical generation and lipid peroxidation. Planta Med 1985; 51(2): 97-100.
[http://dx.doi.org/10.1055/s-2007-969416] [PMID: 4034742]
[95]
Singh H, Kim SJ, Kang DH, et al. Glycyrrhetinic acid as a hepatocyte targeting unit for an anticancer drug delivery system with enhanced cell type selectivity. Chem Commun 2018; 54(87): 12353-6.
[http://dx.doi.org/10.1039/C8CC05175E] [PMID: 30324188]
[96]
Sun Y, Dai C, Yin M, Lu J, Hu H, Chen D. Hepatocellular carcinoma targeted effect of configurations and groups of glycyrrhetinic acid by evaluation of its derivative modified liposomes. Int J Nanomedicine 2018; 13: 1621-32.
[http://dx.doi.org/10.2147/IJN.S153944] [PMID: 29588589]
[97]
Yan T, Cheng J, Liu Z, et al. Acid sensitive polymeric vector targeting to hepatocarcinoma cells via glycyrrhetinic acid receptor mediated endocytosis. Mater Sci Eng C 2018; 87: 32-40.
[http://dx.doi.org/10.1016/j.msec.2018.02.013] [PMID: 29549947]
[98]
Chen J, Chen Y, Cheng Y, Gao Y. Glycyrrhetinic acid liposomes containing mannose-diester lauric diacid cholesterol conjugate synthesized by lipase catalytic acylation for liver-specific delivery. Molecules 2017; 22(10): 1598.
[http://dx.doi.org/10.3390/molecules22101598] [PMID: 28946644]
[99]
Chen G, Li J, Cai Y, et al. A glycyrrhetinic acid modified curcumin supramolecular hydrogel for liver tumor targeting therapy. Sci Rep 2017; 7(1): 44210.
[http://dx.doi.org/10.1038/srep44210] [PMID: 28281678]
[100]
Zhang L, Li J, Chen T, et al. Synthesis, characterization, and in vitro evaluation of curcumin loaded albumin nanoparticles surface-functionalized with glycyrrhetinic acid. Int J Nanomedicine 2015; 10: 5475-87.
[http://dx.doi.org/10.2147/IJN.S88253] [PMID: 26346750]
[101]
Qi WW, Yu HY, Guo H, et al. Doxorubicin loaded glycyrrhetinic acid modified recombinant human serum albumin nanoparticles for targeting liver tumor chemotherapy. Mol Pharm 2015; 12(3): 675-83.
[http://dx.doi.org/10.1021/mp500394v] [PMID: 25584860]
[102]
Tian J, Wang L, Wang L, Ke X, Wogonin LA. A wogonin loaded glycyrrhetinic acid-modified liposome for hepatic targeting with anti tumor effects. Drug Deliv 2014; 21(7): 553-9.
[http://dx.doi.org/10.3109/10717544.2013.853850] [PMID: 24215357]
[103]
Zhang L, Yao J, Zhou J, Wang T, Zhang Q. Glycyrrhetinic acid graft hyaluronic acid conjugate as a carrier for synergistic targeted delivery of antitumor drugs. Int J Pharm 2013; 441(1-2): 654-64.
[http://dx.doi.org/10.1016/j.ijpharm.2012.10.030] [PMID: 23117024]
[104]
Li J, Xu H, Ke X, Tian J. The anti tumor performance of docetaxel liposomes surface modified with glycyrrhetinic acid. J Drug Target 2012; 20(5): 467-73.
[http://dx.doi.org/10.3109/1061186X.2012.685475] [PMID: 22577855]
[105]
Maruyama T, Miyamoto Y, Nakamura T, et al. Identification of membrane-type receptor for bile acids (M-BAR). Biochem Biophys Res Commun 2002; 298(5): 714-9.
[http://dx.doi.org/10.1016/S0006-291X(02)02550-0] [PMID: 12419312]
[106]
Festa C, De Marino S, Carino A, et al. Targeting bile acid receptors: Discovery of a potent and selective farnesoid X receptor agonist as a new lead in the pharmacological approach to liver diseases. Front Pharmacol 2017; 8: 162.
[http://dx.doi.org/10.3389/fphar.2017.00162] [PMID: 28424617]
[107]
Matloub AA, Salama AH, Aglan HA, AbouSamra MM, ElSouda SSM, Ahmed HH. Exploiting bilosomes for delivering bioactive polysaccharide isolated from Enteromorpha intestinalis for hacking hepatocellular carcinoma. Drug Dev Ind Pharm 2018; 44(4): 523-34.
[http://dx.doi.org/10.1080/03639045.2017.1402922] [PMID: 29115890]
[108]
Mohsen AM, Asfour MH, Salama AAA. Improved hepatoprotective activity of silymarin via encapsulation in the novel vesicular nanosystem bilosomes. Drug Dev Ind Pharm 2017; 43(12): 2043-54.
[http://dx.doi.org/10.1080/03639045.2017.1361968] [PMID: 28756693]
[109]
Parashar P, Rana P, Dwivedi M, Saraf SA. Dextrose modified bilosomes for peroral delivery: Improved therapeutic potential and stability of silymarin in diethylnitrosamine-induced hepatic carcinoma in rats. J Liposome Res 2019; 29(3): 251-63.
[http://dx.doi.org/10.1080/08982104.2018.1551408] [PMID: 30501440]
[110]
Scholz C, Wagner E. Therapeutic plasmid DNA versus siRNA delivery: Common and different tasks for synthetic carriers. J Control Release 2012; 161(2): 554-65.
[http://dx.doi.org/10.1016/j.jconrel.2011.11.014] [PMID: 22123560]
[111]
Gary DJ, Puri N, Won YY. Polymer based siRNA delivery: Perspectives on the fundamental and phenomenological distinctions from polymer based DNA delivery. J Control Release 2007; 121(1-2): 64-73.
[http://dx.doi.org/10.1016/j.jconrel.2007.05.021] [PMID: 17588702]
[112]
Zhao L, Li Y, Pei D, et al. Glycopolymers/PEI complexes as serum tolerant vectors for enhanced gene delivery to hepatocytes. Carbohydr Polym 2019; 205: 167-75.
[http://dx.doi.org/10.1016/j.carbpol.2018.10.036] [PMID: 30446092]
[113]
Zamboni CG, Kozielski KL, Vaughan HJ, et al. Polymeric nanoparticles as cancer specific DNA delivery vectors to human hepatocellular carcinoma. J Control Release 2017; 263: 18-28.
[http://dx.doi.org/10.1016/j.jconrel.2017.03.384] [PMID: 28351668]
[114]
Tsuchiya A, Kang JH, Mori T, et al. Efficient delivery of signal responsive gene carriers for disease-specific gene expression via bubble liposomes and sonoporation. Colloids Surf B Biointerfaces 2017; 160: 60-4.
[http://dx.doi.org/10.1016/j.colsurfb.2017.09.021] [PMID: 28917150]
[115]
Khalvati B, Sheikhsaran F, Sharifzadeh S, et al. Delivery of plasmid encoding interleukin-12 gene into hepatocytes by conjugated polyethylenimine based nanoparticles. Artif Cells Nanomed Biotechnol 2017; 45(5): 1036-44.
[http://dx.doi.org/10.1080/21691401.2016.1202256] [PMID: 27357209]
[116]
Sebestyén MG, Wong SC, Trubetskoy V, Lewis DL, Wooddell CI. Targeted in vivo delivery of siRNA and an endosome releasing agent to hepatocytes. Methods Mol Biol 2015; 1218: 163-86.
[http://dx.doi.org/10.1007/978-1-4939-1538-5_10] [PMID: 25319651]
[117]
Petrov RA, Maklakova SY, Ivanenkov YA, et al. Synthesis and biological evaluation of novel mono- and bivalent ASGP-R-targeted drug-conjugates. Bioorg Med Chem Lett 2018; 28(3): 382-7.
[http://dx.doi.org/10.1016/j.bmcl.2017.12.032] [PMID: 29269214]
[118]
Ong ZY, Yang C, Gao SJ, Ke XY, Hedrick JL, Yan Yang Y. Galactose-functionalized cationic polycarbonate diblock copolymer for targeted gene delivery to hepatocytes. Macromol Rapid Commun 2013; 34(21): 1714-20.
[http://dx.doi.org/10.1002/marc.201300538] [PMID: 24123359]
[119]
Rozema DB, Lewis DL, Wakefield DH, et al. Dynamic polyconjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Natl Acad Sci USA 2007; 104(32): 12982-7.
[http://dx.doi.org/10.1073/pnas.0703778104] [PMID: 17652171]
[120]
Kuo TK, Hung SP, Chuang CH, et al. Stem cell therapy for liver disease: Parameters governing the success of using bone marrow mesenchymal stem cells. Gastroenterology 2008; 134(7): 2111-21.
[http://dx.doi.org/10.1053/j.gastro.2008.03.015] [PMID: 18455168]
[121]
Wu HH, Lee OK. Exosomes from mesenchymal stem cells induce the conversion of hepatocytes into progenitor oval cells. Stem Cell Res Ther 2017; 8(1): 117.
[http://dx.doi.org/10.1186/s13287-017-0560-z] [PMID: 28535778]
[122]
Dewangan HK, Pandey T, Maurya L, Singh S. Rational design and evaluation of HBsAg polymeric nanoparticles as antigen delivery carriers. Int J Biol Macromol 2018; 111: 804-12.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.01.073] [PMID: 29343454]
[123]
Tian Y, Liu Z, Zhang L, et al. Combined with ultrasound-targeted nanobubble destruction for synergistic treatment of Hep G2 Cells in vitro. OncoTargets Ther 2018; 11: 4785-95.
[http://dx.doi.org/10.2147/OTT.S170786] [PMID: 30127626]
[124]
Situ JQ, Wang XJ, Zhu XL, et al. Multifunctional SPIO/DOX-loaded A54 homing peptide functionalized dextran-g-PLGA micelles for tumor therapy and MR imaging. Sci Rep 2016; 6(1): 35910.
[http://dx.doi.org/10.1038/srep35910] [PMID: 27775017]
[125]
Wang J, Wang H, Li J, et al. iRGD-decorated polymeric nanoparticles for the efficient delivery of vandetanib to hepatocellular carcinoma: Preparation and in vitro and in vivo evaluation. ACS Appl Mater Interfaces 2016; 8(30): 19228-37.
[http://dx.doi.org/10.1021/acsami.6b03166] [PMID: 27381493]
[126]
Lo A, Lin CT, Wu HC. Hepatocellular carcinoma cell specific peptide ligand for targeted drug delivery. Mol Cancer Ther 2008; 7(3): 579-89.
[http://dx.doi.org/10.1158/1535-7163.MCT-07-2359] [PMID: 18347144]
[127]
Singh V, Garg A, Dewangan HK. Recent advances in drug design and delivery across biological barriers using computational models. Lett Drug Des Discov 2022; 19.
[http://dx.doi.org/10.2174/1570180819999220204110306]
[128]
Chowdhury EH. Nuclear targeting of viral and non-viral DNA. Expert Opin Drug Deliv 2009; 6(7): 697-703.
[http://dx.doi.org/10.1517/17425240903025744] [PMID: 19552613]
[129]
Kaiser RA, Mao SA, Glorioso J, et al. Lentiviral vector-mediated gene therapy of hepatocytes ex vivo for autologous transplantation in swine. J Vis Exp 2018; 141(141)
[http://dx.doi.org/10.3791/58399] [PMID: 30451238]
[130]
Sun Y, Sun Y, Zhao R. Establishment of MicroRNA delivery system by PP7 bacteriophage-like particles carrying cell penetrating peptide. J Biosci Bioeng 2017; 124(2): 242-9.
[http://dx.doi.org/10.1016/j.jbiosc.2017.03.012] [PMID: 28442387]
[131]
Chen BD, He CH, Chen XC, et al. Targeting transgene to the heart and liver with AAV9 by different promoters. Clin Exp Pharmacol Physiol 2015; 42(10): 1108-17.
[http://dx.doi.org/10.1111/1440-1681.12453] [PMID: 26173818]
[132]
Gabriel N, Samuel R, Jayandharan GR. Targeted delivery of AAV transduced mesenchymal stromal cells to hepatic tissue for ex vivo gene therapy. J Tissue Eng Regen Med 2017; 11(5): 1354-64.
[http://dx.doi.org/10.1002/term.2034] [PMID: 26053555]
[133]
Zhang Y, Wei H, Xu L, et al. Preparation and evaluation of a non viral gene vector for SiRNA: Multifunctional envelope-type nano device. Artif Cells Nanomed Biotechnol 2016; 44(5): 1259-65.
[http://dx.doi.org/10.3109/21691401.2015.1024840] [PMID: 25813567]
[134]
Terpstra V, Amersfoort ES, van Velzen AG, Kuiper J, van Berkel TJC. Hepatic and extrahepatic scavenger receptors: Function in relation to disease. Arterioscler Thromb Vasc Biol 2000; 20(8): 1860-72.
[http://dx.doi.org/10.1161/01.ATV.20.8.1860] [PMID: 10938005]
[135]
Rajora MA, Zheng G. Targeting SR-BI for cancer diagnostics, imaging and therapy. Front Pharmacol 2016; 7: 326.
[http://dx.doi.org/10.3389/fphar.2016.00326] [PMID: 27729859]
[136]
Campbell F, Bos FL, Sieber S, et al. Directing nanoparticle biodistribution through evasion and exploitation of stab2-dependent nanoparticle uptake. ACS Nano 2018; 12(3): 2138-50.
[http://dx.doi.org/10.1021/acsnano.7b06995] [PMID: 29320626]
[137]
Liu C, Zhou Z, Chen Y, Liu J, Wang Y, Liu H. Targeted delivery of garcinia glycosides by reconstituted high-density lipoprotein nano complexes. J Microencapsul 2018; 35(2): 115-20.
[http://dx.doi.org/10.1080/02652048.2017.1413146] [PMID: 29195484]
[138]
Svendsen P, Graversen JH, Etzerodt A, et al. Antibody directed glucocorticoid targeting to CD163 in M2-type macrophages attenuates fructose-induced liver inflammatory changes. Mol Ther Methods Clin Dev 2017; 4: 50-61.
[http://dx.doi.org/10.1016/j.omtm.2016.11.004] [PMID: 28344991]
[139]
Vercauteren K, Eede VDN, Mesalam AA, et al. Successful anti scavenger receptor class B type I (SR-BI) monoclonal antibody therapy in humanized mice after challenge with HCV variants with in vitro resistance to SR-BI-targeting agents. Hepatology 2014; 60(5): 1508-18.
[http://dx.doi.org/10.1002/hep.27196] [PMID: 24797654]
[140]
Yuan Y, Wang W, Wang B, Zhu H, Zhang B, Feng M. Delivery of hydrophilic drug doxorubicin hydrochloride targeted liver using apoAI as carrier. J Drug Target 2013; 21(4): 367-74.
[http://dx.doi.org/10.3109/1061186X.2012.757769] [PMID: 23600747]
[141]
Rensen P, Gras JCE, Lindfors E, et al. Selective targeting of liposomes to macrophages using a ligand with high affinity for the macrophage scavenger receptor class A. Curr Drug Discov Technol 2006; 3(2): 135-44.
[http://dx.doi.org/10.2174/157016306778108893] [PMID: 16925521]
[142]
Zhou Q, Sun X, Zeng L, Liu J, Zhang Z. A randomized multicenter phase II clinical trial of mitoxantrone loaded nanoparticles in the treatment of 108 patients with unresected hepatocellular carcinoma. Nanomedicine 2009; 5(4): 419-23.
[http://dx.doi.org/10.1016/j.nano.2009.01.009] [PMID: 19523421]
[143]
Kiselev MA, Selyakov DN, Gapon IV, et al. Investigation of nanodrug phospholipovit by small angle neutron scattering. Crystallogr Rep 2019; 64(4): 656-61.
[http://dx.doi.org/10.1134/S1063774519040114]
[144]
Kudinov VA, Ipatova OM, Fyodorov IG, et al. Investigation of efficacy of phospholipovit for correction of the hepatic encephalopathy. Biomed Khim 2016; 62(6): 704-7.
[http://dx.doi.org/10.18097/PBMC20166206704] [PMID: 28026815]
[145]
Siqing F, Culotta KS, Falchook GS, et al. Pharmacokinetic evaluation of nanoparticle albumin bound paclitaxel delivered via hepatic arterial infusion in patients with predominantly hepatic metastases. Cancer Chemother Pharmacol 2016; 77(2): 357-64.
[http://dx.doi.org/10.1007/s00280-015-2946-x] [PMID: 26698868]
[146]
Nathwani AC, Tuddenham EGD, Rangarajan S, et al. Adenovirus associated virus vector mediated gene transfer in hemophilia B. N Engl J Med 2011; 365(25): 2357-65.
[http://dx.doi.org/10.1056/NEJMoa1108046] [PMID: 22149959]
[147]
Kehagias DT, Gouliamos AD, Smyrniotis V, Vlahos LJ. Diagnostic efficacy and safety of MRI of the liver with superparamagnetic iron oxide particles (SH U 555 A). J Magn Reson Imaging 2001; 14(5): 595-601.
[http://dx.doi.org/10.1002/jmri.1224] [PMID: 11747012]
[148]
Wu TS. Medicinal herbal composition for treating liver diseases and HIV US Patent 6455078B1, 2002.
[149]
Mitra S, Saxena E, Dixit M. Natural composition for curing hepatitis-B, methods for making the same and pharmaceutical formulations thereof US Patent 20060110479A1, 2006.
[150]
Jalan R, Jalan KN. Compositions comprising ornithine and phenylacetate or phenylbutyrate for treating hepatic encephalopathy US Patent 10610506, 2020.
[151]
Aleksandrova K, Pediaditakis P, Salisbury J, Rüdinger W. Encapsulated liver cell composition US Patent 20169517247B2, 2016.
[152]
Howell CA. Porous carbon particles for use in the treatment or prevention of liver disease US Patent 20150064256A1, 2015.
[153]
Davies N, Jalan R. Therapy for liver disease US Patent US8480607B2, 2013.
[154]
Jalan R, Mookerjee RP, Davies N. Biomarkers for assessing liver function US Patent US20090280519A1, 2013.
[155]
Gedulin B, Grey M, O’Donnell N. Bile acid recycling inhibitors for treatment of pediatric cholestatic liver diseases US Patent US20130109671A1, 2013.
[156]
Zabrecky G. Methods and formulations for treating chronic liver disease US Patent US8197861B2, 2012.
[157]
Bok SH, Jeong TS, Bae KH, et al. Method for preventing or treating elevated blood lipid level related diseases by administering natural phenolic compounds US Patent US6133311A1, 2000.
[158]
Karmali RA. Salts of aminoimidazole carboxamide useful in the prevention and treatment of liver diseases US Patent US6184227B1, 2001.
[159]
Gillberg PG, Graffner H, Starke I. IBAT inhibitors for the treatment of liver diseases US Patent US9688720B2, 2017.
[160]
Frohlich E, Kvietikova I, Zatloukal K, et al. Use of mitochondrially targeted antioxidant in the treatment of liver diseases and epithelial cancers US Patent 20070225255A1, 2007.
[161]
Egashira K, Takeshita A, Koyanagi M, Nakamuta M, Nishida KI. Drugs for liver diseases US Patent US7335647B2, 2008.
[162]
Hinkle G, Kotelianski V, Bettencourt B, Sehgal A, Novobrantseva T. Compositions and methods directed to treating liver fibrosis US Patent US10669303B2, 2020.
[163]
Fitzgerald K, Hinkle G. 3 (PNPLA3) iRNA compositions and methods of use thereof US Patent US10231988B2, 2019.
[164]
Guild BC, DeRosa F, Heartlein M. Liver specific delivery of messenger RNA US Patent US10143758B2, 2018.
[165]
Koncarevic A, Lachey J, Seehra J, Sherman ML. Methods for treating fatty liver disease US Patent US20190062392A1, 2019.
[166]
Gonzalez FJ, Jiang C, Xie C, et al. Inhibitors of the farnesoid X receptor and uses in medicine US Patent 10233209B2, 2019.
[167]
Barker DK. Nanoves : Treatment, biomarkers and diagnostic tests for liver diseases and comorbid diseases US Patent US20159186343B2, 2015.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy