1,132
Views
88
CrossRef citations to date
0
Altmetric
Review

Bile acids and bile acid derivatives: use in drug delivery systems and as therapeutic agents

, , &
Pages 1133-1148 | Received 22 Jan 2016, Accepted 06 Apr 2016, Published online: 02 May 2016

References

  • Maldonado-Valderrama J, Wilde P, Macierzanka A, et al. The role of bile salts in digestion. Adv Colloid Interface Sci. 2011;165:36–46.
  • Monte MJ, Marin JJG, Antelo A, et al. Bile acids: chemistry, physiology, and pathophysiology. World J Gastroenterol. 2009 Feb 21;15(7):804–816.
  • Pols TWH, Noriega LG, Nomura M, et al. The bile acid membrane receptor TGR5 as an emerging target in metabolism and inflammation. J Hepatol. 2011;54:1263–1272.
  • Keitel V, Kubitz R, Häussinger D. Endocrine and paracrine role of bile acids. World J Gastroenterol. 2008;14:5620–5629.
  • Quinn M, DeMorrow SB. Bile in the brain? A role for bile acids in the central nervous system. J Cell Sci Ther. 2012;3:7.
  • Hofmann AF, Hagey LR. Bile acids: chemistry, pathochemistry, biology, pathobiology, and therapeutics. Cell Mol Life Sci. 2008;65:2461–2483.
  • Alrefai WA, Gill RK. Bile acid transporters: structure, function, regulation and pathophysiological implications. Pharm Res. 2007;24:1803–1823.
  • Sievänen E. Exploitation of bile acid transport systems in prodrug design. Molecules. 2007;12:1859–1889.
  • Balakrishnan A, Polli JE. Apical sodium dependent transporter (ASBT, SLC10A2): a potential prodrug target. Mol Pharm. 2006;3:223–230.
  • Kramer W, Glombik H. Bile acid reabsorption inhibitors (BARI): novel hypolipidemic drugs. Curr Med Chem. 2006;13:997–1016.
  • Madenci D, Egelhaaf SU. Self-assembly in aqueous bile salt solutions. J Colloid Interface Sci. 2010;15:109–115.
  • Calabresi M, Andreozzi P, La Mesa C. Supramolecular association and polymorphic behaviour in systems containing bile acid salts. Molecules. 2007;12:1731–1754.
  • Natalini B, Sardella R, Gioiello A, et al. Determination of bile salt critical micellization concentration on the road to drug discovery. J Pharm Biomed Anal. 2014;87:62–81.
  • Nonnappa MU Unlocking the potential of bile acids in synthesis, supramolecular/materials chemistry and nanoscience. Org Biomol Chem. 2008;6:657–669.
  • Pártay LB, Sega M, Jedlovszky P. Morphology of bile salt micelles as studied by computer simulation methods. Langmuir. 2007;23:12322–12328.
  • Faustino C, Serafim C, Ferreira I, et al. Solubilisation power of an amino acid-based gemini surfactant towards the hydrophobic drug amphotericin B. Colloids Surf A. 2014;480:426–432.
  • Serafim C, Ferreira I, Rijo P, et al. Lipoamino acid-based micelles as promising delivery vehicles for monomeric amphotericin B. Int J Pharm. 2015;497:23–35.
  • Galantini L, di Gregorio CM, Gubitosi M, et al. Bile salts and derivatives: rigid unconventional amphiphiles as dispersants, carriers and superstructure building blocks. Curr Opin Colloid Interface Sci. 2015;20:170–182.
  • di Gregorio CM, Pavel NV, Jover A, et al. pH sensitive tubules of a bile acid derivative: a tubule opening by release of wall leaves. Phys Chem Chem Phys. 2013;15:7560–7566.
  • Natalini B, Sardella R, Camaioni E, et al. Correlation between CMC and chromatographic index: simple and effective evaluation of the hydrophobic/hydrophilic balance of bile acids. Anal Bioanal Chem. 2007;388:1681–1688.
  • Roda A, Minutello A, Angellotti MA, et al. Bile acid structure-activity relationship: evaluation of bile acid lipophilicity using 1-octanol/water partition coefficient and reverse phase HPLC. J Lipid Res. 1990;31:1433–1443.
  • Stojancevic M, Pavlovic N, Golocorbin-Kon S, et al. Application of bile acids in drug formulation and delivery. Front Life Sci. 2013;7:112–122.
  • Garidel P, Hildebrand A, Knauf K, et al. Membranolytic activity of bile salts: influence of biological membrane properties and composition. Molecules. 2007;12:2292–2326.
  • Moghimipour E, Ameri A, Handali S. Absorption-enhancing effects of bile salts. Molecules. 2015;20:14451–14473.
  • Patra D, Ahmadieh D, Aridi R. Study on interaction of bile salts with curcumin and curcumin embedded in dipalmitoyl-sn-glycero-3-phosphocholine liposome. Colloids Surf B. 2013;110:296–304.
  • Zhou Y, Maxwell KN, Sezgin E, et al. Bile acids modulate signalling by functional perturbation of plasma membrane domains. J Biol Chem. 2013;288:35660–35670.
  • Sharma R, Majer F, Peta VK, et al. Bile acid toxicity structure-activity relationships: correlations between cell viability and lipophilicity in a panel of new and known bile acids using an oesophageal cell line (HET-1A). Bioorg Med Chem. 2010;18:6886–6895.
  • Singh M, Singh A, Kundu S, et al. Deciphering the role of charge, hydration, and hydrophobicity for cytotoxic activities and membrane interactions of bile acid based facial amphiphiles. Biochim Biophys Acta. 2013;1828:1926–1937.
  • Randazzo RAS, Bucki R, Janmey PA, et al. A series of cationic sterol lipids with gene transfer and bactericidal activity. Bioorg Med Chem. 2009;17:3257–3265.
  • Bhattacharjee J, Verma G, Aswal VK, et al. Tween 80-sodium deoxycholate mixed micelles: structural characterization and application in doxorubicin delivery. J Phys Chem B. 2010;114:16414–16421.
  • Tan Y, Qi J, Lu Y, et al. Lecithin in mixed micelles attenuates the cytotoxicity of bile salts in Caco-2 cells. Toxicol In Vitro. 2013;27:714–720.
  • Faustino CMC, Serafim CS, Ferreira IN, et al. Mixed micelle formation between an amino acid-based anionic gemini surfactant and bile salts. Ind Eng Chem Res. 2014;53:10112–10118.
  • Maswal M, Dar AA. Mixed micelles of sodium cholate and Brij30: their rheological behaviour and capability towards solubilisation and stabilization of rifampicin. Colloids Surf A. 2013;436:704–713.
  • Aburahma MH. Bile salts-containing vesicles: promising pharmaceutical carriers for oral delivery of poorly water-soluble drugs and peptide/protein-based therapeutics or vaccines. Drug Deliv. 2015;8:1–21.
  • Pinto Reis C, Silva C, Martinho N, et al. Drug carriers for oral delivery of peptides and proteins: accomplishments and future perspectives. Ther Deliv. 2013;4:251–265.
  • Pinto Reis C, Neufeld RJ, Ribeiro AJ, et al. Nanoencapsulation II. Biomedical applications and current status of peptide and protein nanoparticulate delivery systems. Nanomedicine. 2006;2:53–65.
  • Shukla A, Khatri K, Gupta PN, et al. Oral immunization against hepatitis B using bile salt stabilized vesicles (bilosomes). J Pharm Pharm Sci. 2008;11:59–66.
  • Shukla A, Katare OP, Singh B, et al. M-cell targeted delivery of recombinant hepatitis B surface antigen using cholera toxin B subunit conjugated bilosomes. Int J Pharm. 2010;385:47–52.
  • Jain S, Harde H, Indulkar A, et al. Improved stability and immunological potential of tetanus toxoid containing surface engineered bilosomes following oral administration. Nanomedicine. 2014;10:431–440.
  • Damgé C, Reis CP, Maincent P. Nanoparticle strategies for the oral delivery of insulin. Expert Opin Drug Del. 2008;5:1–24.
  • Niu M, Lu Y, Hovgaard L, et al. Hypoglycemic activity and oral bioavailability of insulin-loaded liposomes containing bile salts in rats: the effect of cholate type, particle size and administered dose. Eur J Pharm Biopharm. 2012;81:265–272.
  • Song KH, Chung SJ, Shim CK. Enhanced intestinal absorption of salmon calcitonin (sCT) from proliposomes containing bile salts. J Control Release. 2005;106:298–308.
  • Arzani G, Haeri A, Daeihamed M, et al. Niosomal carriers enhance oral bioavailability of carvedilol: effects of bile salt-enriched vesicles and carrier surface charge. Int J Nanomedicine. 2015;10:4797–4813.
  • Al-Mahallawi AM, Abdelbary AA, Aburahma MH. Investigating the potential of employing bilosomes as a novel vesicular carrier for transdermal delivery of tenoxicam. Int J Pharm. 2015;485:329–340.
  • Pinto Reis C, Neufeld RJ, Ribeiro AJ, et al. Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles. Nanomedicine. 2006;2:8–21.
  • Elnaggar YS. Multifaceted applications of bile salts in pharmacy: an emphasis on nanomedicine. Int J Nanomedicine. 2015;10:3955–3971.
  • Shi Z, Guo R, Li W, et al. Nanoparticles of deoxycholic acid, polyethylene glycol and folic acid-modified chitosan for targeted delivery of doxorubicin. J Mater Sci Mater Med. 2014;25:723–731.
  • Xiao K, Li Y, Lee J, et al. “OA02” peptide facilitates the precise targeting of paclitaxel-loaded micellar nanoparticles to ovarian cancer in vivo. Cancer Res. 2012;72:2100–2110.
  • Le Dévédec F, Fuentealba D, Strandman S, et al. Aggregation behavior of pegylated bile acid derivatives. Langmuir. 2012;28:13431–13440.
  • Le Dévédec F, Strandman S, Hildgen P, et al. PEGylated bile acids for use in drug delivery systems: enhanced solubility and bioavailability of itraconazole. Mol Pharm. 2013;10:3057–3066.
  • Zhou H, Yu W, Guo X, et al. Synthesis and characterization of amphiphilic glycidol-chitosan-deoxycholic acid nanoparticles as a drug carrier for doxorubicin. Biomacromolecules. 2010;11:3480–3486.
  • Yan JK, Ma HL, Chen X, et al. Self-aggregated nanoparticles of carboxylic curdlan-deoxycholic acid conjugates as a carrier of doxorubicin. Int J Biol Macromol. 2015;72:333–340.
  • Li J, Huo M, Wang J, et al. Redox-sensitive micelles self-assembled from amphiphilic hyaluronic acid-deoxycholic acid conjugates for targeted intracellular delivery of paclitaxel. Biomaterials. 2012;33:2310–2320.
  • Li J, Yin T, Wang L, et al. Biological evaluation of redox-sensitive micelles based on hyaluronic acid-deoxycholic acid conjugates for tumor-specific delivery of paclitaxel. Int J Pharm. 2015;483:38–48.
  • Chen W-Q, Wei H, Li S-L, et al. Fabrication of star-shaped, thermo-sensitive poly(N-isopropylacrylamide)–cholic acid–poly(ε-caprolactone) copolymers and their self-assembled micelles as drug carriers. Polymer. 2008;49:3965–3972.
  • Tang X, Cai S, Zhang R, et al. Paclitaxel-loaded nanoparticles of star-shaped cholic acid-core PLA-TPGS copolymer for breast cancer treatment. Nanoscale Res Lett. 2013;8:420.
  • Zeng X, Tao W, Mei L, et al. Cholic acid-functionalized nanoparticles of star-shaped PLGA-vitamin E TPGS copolymer for docetaxel delivery to cervical cancer. Biomaterials. 2013;34:6058–6067.
  • Xiao K, Luo J, Fowler WL, et al. A self- assembling nanoparticle for paclitaxel delivery in ovarian cancer. Biomaterials. 2009;30:6006–6016.
  • Li Y, Xiao K, Luo J, et al. A novel size-tunable nanocarrier system for targeted anticancer drug delivery. J Control Release. 2010;144:314–323.
  • Xiao K, Suby N, Li Y, et al. Telodendrimer-based nanocarriers for the treatment of ovarian cancer. Ther Deliv. 2013;4:1279–1292.
  • Xiao K, Li Y, Luo J, et al. The effect of surface charge on in vivo biodistribution of PEG-oligocholic acid based micellar nanoparticles. Biomaterials. 2011;32:3435–3446.
  • Li Y, Xiao K, Luo J, et al. Well-defined, reversible disulfide cross-linked micelles for on-demand paclitaxel delivery. Biomaterials. 2011;32:6633–6645.
  • Li Y, Xiao W, Xiao K, et al. Well-defined, reversible boronate crosslinked nanocarriers for targeted drug delivery in response to acidic pH values and cis-diols. Angew Chem Int Ed Engl. 2012;51:2864–2869.
  • Huang W, Shi C, Shao Y, et al. The core-inversible micelles for hydrophilic drug delivery. Chem Commun (Camb). 2013;49:6674–6676.
  • Kramer W. Transporters, Trojan horses and therapeutics: suitability of bile acid and peptide transporters for drug delivery. Biol Chem 2011;392:77–94.
  • Tolle-Sander S, Lentz KA, Maeda DY, et al. Increased acyclovir oral bioavailability via a bile acid conjugate. Mol Pharm. 2004;1:40–48.
  • Rais R, Acharya C, Mackerell AD, et al. Structural determinants for transport across the intestinal bile acid transporter using C-24 bile acid conjugates. Mol Pharm. 2010;7:2240–2254
  • Lee S, Kim K, Kumar TS, et al. Synthesis and biological properties of insulin-deoxycholic acid chemical conjugates. Bioconjug Chem. 2005;16:615–620.
  • Chen D, Wang X, Chen L, et al. Novel liver-specific cholic acid-cytarabine conjugates with potent antitumor activities: synthesis and biological characterization. Acta Pharmacol Sin. 2011;32:664–672.
  • Wu D, Ji S, Wu Y, et al. Design, synthesis, and antitumor activity of bile acid-polyamine-nucleoside conjugates. Bioorg Med Chem Lett. 2007;17:2983–2986.
  • Qian S, Wu J-B, Wu X-C, et al. Synthesis and characterization of new liver targeting 5-fluorouracil-cholic acid conjugates. Arch Pharm (Weinheim). 2009;342:513–520.
  • Sreekanth V, Bansal S, Motiani RK, et al.. Design, synthesis, and mechanistic investigations of bile acid-tamoxifen conjugates for breast cancer therapy. Bioconjug Chem. 2013;24:1468–1484.
  • Monte MJ, Ballestero MR, Briz O, et al. Proapoptotic effect on normal and tumor intestinal cells of cytostatic drugs with enterohepatic organotropism. J Pharmacol Exp Ther. 2005;315:24–35.
  • Briz O, Macias RIR, Vallejo M, et al. Usefulness of liposomes loaded with cytostatic bile acid derivatives to circumvent chemotherapy resistance of enterohepatic tumors. Mol Pharm. 2003;63:742–750.
  • Anelli PL, Lattuada L, Lorusso V, et al. Conjugates of gadolinium complexes to bile acids as hepatocyte-directed contrast agents for magnetic resonance imaging. J Med Chem. 2004;47:3629–3641.
  • Janout V, Regen SL. Bioconjugate-based molecular umbrellas. Bioconjug Chem. 2009;20:183–192.
  • Janout V, Schell WA, Thévenin D, et al. Taming amphotericin B. Bioconjug Chem. 2015;26:2021–2024.
  • Ikegami T, Matsuzaki Y. Ursodeoxycholic acid: mechanism of action and novel clinical applications. Hepatol Res. 2008;38:123–131.
  • Bernstein H, Bernstein C, Payne CM, et al. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 2009;15:3329–3340.
  • Kundu S, Kumar S, Bajaj A. Cross-talk between bile acids and gastrointestinal tract for progression and development of cancer and its therapeutic implications. IUBMB Life. 2015;67:514–523.
  • Horowitz NS, Hua J, Powell MA, et al. Novel cytotoxic agents from an unexpected source: bile acids and ovarian tumor apoptosis. Gynecol Oncol. 2007;107:344–349.
  • Kim ND, Im E, Yoo YH, et al. Modulation of the cell cycle and induction of apoptosis in human cancer cells by synthetic bile acids. Curr Cancer Drug Targets. 2006;6:681–689.
  • Agarwal DS, Anantaraju HS, Sriram D, et al. Synthesis, characterization and biological evaluation of bile acid-aromatic/heteroaromatic amides linked via amino acids as anti-cancer agents. Steroids. 2016;107:87–97.
  • Brossard D, Lechevrel M, El Kihel L, et al. Synthesis and biological evaluation of bile carboxamide derivatives with pro-apoptotic effect on human colon adenocarcinoma cell lines. Eur J Med Chem. 2014;86:279–290.
  • Chong H-S, Song HA, Ma X, et al. Bile acid-based polyaminocarboxylate conjugates as targeted antitumor agents. Chem Commun (Camb). 2009 Jun 7;(21):3011–3013. doi: 10.1039/b823000e.
  • Vallejo M, Castro MA, Medarde M, et al. Novel bile acid derivatives (BANBs) with cytostatic activity obtained by conjugation of their side chain with nitrogenated bases. Biochem Pharmacol. 2007;73:1394–1404.
  • Xu Y, Luo Q, Lin T, et al. U12, a UDCA derivative, acts as an anti-hepatoma drug lead and inhibits the mTOR/S6K1 and cyclin/CDK complex pathways. PLoS One. 2014;9:e113479.
  • Singh M, Bansal S, Kundu S, et al. Synthesis, structure-activity relationship, and mechanistic investigation of lithocholic acid amphiphiles for colon cancer therapy. Med Chem Commun. 2015;6:192–201.
  • Popadyuk II, Markov AV, Salomatina OV, et al. Synthesis and biological activity of novel deoxycholic acid derivatives. Bioorg Med Chem 2015;23:5022–5034.
  • Vang S, Longley K, Steer CJ, et al. The unexpected uses of urso- and tauroursodeoxycholic acid in the treatment of non-liver diseases. Glob Adv Health Med. 2014;3:58–69.
  • Amaral JD, Viana RJ, Ramalho RM, et al. Bile acids: regulation of apoptosis by ursodeoxycholic acid. J Lipid Res. 2009;50:1721–1734.
  • Palmela I, Correia L, Silva RFM, et al. Hydrophilic bile acid protect human blood-brain barrier endothelial cells from disruption by unconjugated bilirubin: an in vitro study. Front Neurosci. 2015;9:80.
  • Ramalho RM, Viana RJ, Low WC, et al. Bile acids and apoptosis modulation: an emerging role in experimental Alzheimer’s disease. Trends Mol Med. 2008;14:54–62.
  • Lo AC, Callaerts-Vegh Z, Nunes AF, et al. Tauroursodeoxycholic acid (TUDCA) supplementation prevents cognitive impairment and amyloid deposition in APP/PS1 mice. Neurobiol Dis. 2013;50:21–29.
  • Cortez LM, Campeau J, Norman G, et al. Bile acids reduce prion conversion, reduce neuronal loss, and prolong male survival in models of prion disease. J Virol. 2015;89:7660–7672.
  • Lai XZ, Feng Y, Pollard J, et al. Ceragenins: cholic acid-based mimics of antimicrobial peptides. Acc Chem Res. 2008;41:1233–1240.
  • Niemirowicz K, Surel U, Wilczewska AZ, et al. Bactericidal activity and biocompatibility of ceragenin-coated magnetic nanoparticles. J Nanobiotechnology 2015;13:32.
  • Epand RF, Pollard JE, Wright JO, et al. Depolarization, bacterial membrane composition, and the antimicrobial action of ceragenins. Antimicrob Agents Chemother. 2010;54:3708–3713.
  • Bansal S, Singh M, Kidwai S, et al. Bile acid amphiphiles with tunable head groups as highly selective antitubercular agents. Med Chem Commun. 2014;5:1761–1768.
  • Pore VS, Divse JM, Charolkar CR, et al. Design and synthesis of 11α-substituted bile acid derivatives as potential anti-tuberculosis agents. Bioorg Med Chem Lett. 2015;25:4185–4190.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.