332
Views
9
CrossRef citations to date
0
Altmetric
Reviews

Formulations of deoxycholic for therapy: a patent review (2011 – 2014)

, &

Bibliography

  • Papers of special note have been highlighted as either of interest (*) or of considerable interest (**) to readers.
  • Russell DW. The enzymes, regulation, and genetics of bile acid synthesis. Annu Rev Biochem. 2003;72:137–174.

**A comprehensive review of chemical and biological background

**A report focused on the anti-cancer activity of steroids

*A comprehensive review of biological effects of bile acids (BAs) on cancer diseases

  • 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.
  • Baptissart M, Vega A, Maqdasy S, et al. Bile acids: from digestion to cancers. Biochimie. 2013;95:504–517.

*A comprehensive review of biological effects of BAs on cancer diseases

  • Im EO, Choi YH, Paik KJ, et al. Novel bile acid derivatives induce apoptosis via a p53-independent pathway in human breast carcinoma cells. Cancer Lett. 2001;163:83–93.
  • Choi YH, Im EO, Suh H, et al. Apoptotic activity of novel bile acid derivatives in human leukemic T cells through the activation of caspases. Int J Oncol. 2001;18:979–984.
  • Jeong JH, Park JS, Moon B, et al. Orphan nuclear receptor Nur77 translocates to mitochondria in the early phase of apoptosis induced by synthetic chenodeoxycholic acid derivatives in human stomach cancer cell line SNU-1. Ann N Y Acad Sci. 2003;1010:171–177.
  • Choi YH, Im EO, Suh H, et al. Apoptosis and modulation of cell cycle control by synthetic derivatives of ursodeoxycholic acid and chenodeoxycholic acid in human prostate cancer cells. Cancer Lett. 2003;199:157–167.
  • Im EO, Choi SH, Suh H, et al. Synthetic bile acid derivatives induce apoptosis through a c-Jun N-terminal kinase and NF-kappaB-dependent process in human cervical carcinoma cells. Cancer Lett. 2005;229:49–57.
  • Yee SB, Yeo WJ, Park BS, et al. Synthetic chenodeoxycholic acid derivatives inhibit glioblastoma multiform tumor growth in vitro and in vivo. Int J Oncol. 2005;27:653–659.
  • 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.
  • Park SE, Lee SW, Hossain MA, et al. A chenodeoxycholic derivative, HS-1200, induces apoptosis and cell cycle modulation via Egr-1 gene expression control on human hepatoma cells. Cancer Lett. 2008;270:77–86.
  • Asciutti S, Castellani D, Nardi E, et al. A new amino acid derivative of ursodeoxycholate, (N-L-Glutamyl)-UDCA (UDCA-Glu), to selectively release UDCA in the colon. Anticancer Res. 2009;29:4971–4979.
  • Lichtenberg D, Robson RJ, Dennis EA. Solubilization of phospholipids by detergents. Structural and kinetic aspects. Biochim Biophys Acta. 1983;737:285–304.
  • Alkan-Onyuksel H, Ramakrishnan S, Chai HB, et al. A mixed micellar formulation suitable for the parenteral administration of taxol. Pharm Res. 1994;11:206–212.
  • Hammad MA, Muller BW. Increasing drug solubility by means of bile salt-phosphatidylcholine-based mixed micelles. Eur J Pharm Biopharm. 1998;46:361–367.
  • Durr M, Hager J, Lohr JP. Investigation on mixed micelle and liposome preparations for parental use on soya phosphatidylcholine. Eur J Pharm Biopharm. 1994;40:147–156.
  • Rzany B, Griffiths T, Walker P, et al. Reduction of unwanted submental fat with ATX-101 (deoxycholic acid), an adipocytolytic injectable treatment: results from a phase III, randomized, placebo-controlled study. Br J Dermatol. 2014;170:445–453.

*Results of a phase III clinical study for evaluation of the efficacy and safety of ATX-101 treatment for the reduction of submental fat (SMF)

  • Chen X, Oshima T, Tomita T, et al. Acidic bile salts modulate the squamous epithelial barrier function by modulating tight junction proteins. Am J Physiol Gastrointest Liver Physiol. 2011;301:G203–G209.
  • Jeon OC, Hwang SR, Al-Hilal TA, et al. Oral delivery of ionic complex of ceftriaxone with bile acid derivative in non-human primates. Pharm Res. 2013;30:959–967.
  • Balakrishnan A, Polli JE. Apical sodium dependent bile acid transporter (ASBT, SLC10A2): a potential prodrug target. Mol Pharm. 2006;3:223–230.
  • Zhu Y, Yu J, Tong S, et al. Preparation and in vitro evaluation of povidone-sodium cholate-phospholipid mixed micelles for the solubilization of poorly soluble drugs. Arch Pharm Res. 2010;33(6):911–917.
  • Rittes PG. The use of phosphatidylcholine for correction of lower lid bulging due to prominent fat pads. Dermatol Surg. 2001;27:391–392.
  • Duncan DI, Hasengschwandtner F. Lipodissolve for subcutaneous fat reduction and skin retraction. Aesthet Surg J. 2005;25:530–543.
  • Bechara FG, Sand M, Sand D, et al. Lipolysis of lipomas in patients with familial multiple lipomatosis: an ultrasonography-controlled trial. J Cutan Med Surg. 2006;10:155–159.
  • Kutlubay Z. Evaluation of mesotherapeutic injections of three different combinations of lipolytic agents for body contouring. J Cosmet Laser Ther. 2011;13:142–153.
  • Hunter NS, El Tawdy AM, Hegazy RA, et al. A 15-patient pilot trial of lipolysis of the hips and thighs using a phosphatidylcholine and deoxycholate formulation. Dermatol Surg. 2013;39:791–794.
  • Klein SM, Schreml S, Nerlich M, et al. In vitro studies investigating the effect of subcutaneous phosphatidylcholine injections in the 3T3-L1 adipocyte model: lipolysis or lipid dissolution? Plast Reconstr Surg. 2009;124:419–427.
  • Rotunda AM, Kolodney MS. Mesotherapy and phosphatidylcholine injections: historical clarification and review. Dermatol Surg. 2006;32:465–480.
  • Bechara FG, Mannherz HG, Jacob M, et al. Induction of fat cell necrosis in human fat tissue after treatment with phosphatidylcholine and deoxycholate. J Eur Acad Dermatol Venereol. 2012;26:180–185.
  • Kopera D, Binder B, Toplak H, et al. Histopathologic changes after intralesional application of phosphatidylcholine for lipoma reduction: report of a case. Am J Dermatopathol. 2006;28:331–333.
  • Rotunda AM, Ablon G, Kolodney MS. Lipomas treated with subcutaneous deoxycholate injections. J Am Acad Dermatol. 2005;53:973–978.
  • Bechara FG, Sand M, Hoffmann K, et al. Fat tissue after lipolysis of lipomas: a histopathological and immunohistochemical study. J Cutan Pathol. 2007;34:552–557.
  • Palmer M, Curran J, Bowler P. Clinical experience and safety using phosphatidylcholine injections for the localized reduction of subcutaneous fat: a multicentre, retrospective UK study. J Cosmet Dermatol. 2006;5:218–226.
  • US Food and Drug Administration. FDA issues warning letters for drugs promoted in fat elimination procedure. FDA News Release [Internet]. 2010 April 7 [cited 2015 Jul 15]. Available from: http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2010/ucm207453.htm.
  • Non-surgical lipolysis banned in France [cited 2015 July 15]. Available from: http://www.consultingroom.com/blog/184/non-surgical-lipolysis-banned-in-france.
  • Rotunda AM, Suzuki H, Moy RL, et al. Detergent effects of sodium deoxycholate are a major feature of an injectable phosphatidylcholine formulation used for localized fat dissolution. Dermatol Surg. 2004;30:1001–1008.
  • Kolodney MS, Rotunda AM. Method and related compositions for reduction of fat. US2011002896A1. 2011.
  • Kolodney MS, Rotunda AM. Methods and related compositions for reductions of fat and skin tightening. US20140004206A1. 2014.
  • Kythera Biopharmaceuticals, Inc. Formulations of deoxycholic acid and salts thereof. US20120258943A1. 2012.
  • Walker P. Treatment of submental fat. US20120237492A1. 2012.
  • Ascher B, Hoffmann K, Walker P, et al. Efficacy, patient-reported outcomes and safety profile of ATX-101 (deoxycholic acid), an injectable drug for the reduction of unwanted submental fat: results from a phase III, randomized, placebo-controlled study. J Eur Acad Dermatol Venereol. 2014;28:1707–1715.

**Results of a phase III clinical study for evaluation of the efficacy and safety of ATX-101 treatment for the reduction of SMF

  • Walker P, Fellmann J, Lizzul PF. A phase I safety and pharmacokinetic study of ATX-101: injectable, synthetic deoxycholic acid for submental contouring. J Drugs Dermatol. 2015;14(3):279–287.

**Results of a phase I clinical study of ATX-101

  • Walker P, Lee D. A phase 1 pharmacokinetic study of ATX-101: serum lipids and adipokines following synthetic deoxycholic acid injections. J Cosmet Dermatol. 2015;14:33–39.

**Results of a phase I clinical study of ATX-101

  • Schuller-Petrovic S, Wolkart G, Hofler G, et al. Tissue-toxic effects of phosphatidylcholine/deoxycholate after subcutaneous injection for fat dissolution in rats and a human volunteer. Dermatol Surg. 2008;34:529–542.
  • Gupta A, Lobocki C, Singh S, et al. Actions and comparative efficacy of phosphatidylcholine formulation and isolated sodium deoxycholate for different cell types. Aesthetic Plast Surg. 2009;33:346–352.
  • Duncan D, Golitz L Tissue toxic effects of sodium deoxycholate dominant formulas for injection lipolysis. Pre-sented at: FSPS Annual Meeting; 2008 Dec 5; Palm Beach, FL.
  • Wollina U, Goldman A. ATX-101 for reduction of submental fat. Expert Opin Pharmacother. 2015;16:755–762.
  • Centuori SM, Martinez JD. Differential regulation of EGFR-MAPK signaling by deoxycholic acid (DCA) and ursodeoxycholic acid (UDCA) in colon cancer. Dig Dis Sci. 2014;59:2367–2380.

*A comprehensive review of biological effects of deoxycholic acid (DOCA) on colon cancer development

  • Bernstein C, Bernstein H, Garewal H, et al. A bile acid-induced apoptosis assay for colon cancer risk and associated quality control studies. Cancer Res. 1999;59:2353–2357.
  • Bernstein C, Holubec H, Bhattacharyya AK, et al. Carcinogenicity of deoxycholate, a secondary bile acid. Arch Toxicol. 2011;85:863–871.
  • Powell AA, LaRue JM, Batta AK, et al. Bile acid hydrophobicity is correlated with induction of apoptosis and/or growth arrest in hct116 cells. Biochem J. 2001;356:481–486.
  • Jean-Louis S, Akare S, Ali MA, et al. Deoxycholic acid induces intracellular signaling through membrane perturbations. J Biol Chem. 2006;281:14948–14960.
  • Powell AA, Akare S, Qi W, et al. Resistance to ursodeoxycholic acid-induced growth arrest can also result in resistance to deoxycholic acid-induced apoptosis and increased tumorgenicity. BMC Cancer. 2006;6:219.
  • Rosignoli P, Fabiani R, De Bartolomeo A, et al. Genotoxic effect of bile acids on human normal and tumour colon cells and protection by dietary antioxidants and butyrate. Eur J Nutr. 2008;47:301–309.
  • Ignacio Barrasa J, Olmo N, Pérez-Ramos P, et al. Deoxycholic and chenodeoxycholic bile acids induce apoptosis via oxidative stress in human colon adenocarcinoma cells. Apoptosis. 2011;16:1054–1067.
  • Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis. Eur J Cancer. 2001;37:S9–15.
  • Qiao D, Chen W, Stratagoules ED, et al. Bile acidinduced activation of activator protein-1 requires both extracellular signal-regulated kinase and protein kinase c signaling. J Biol Chem. 2000;275:15090–15098.
  • Qiao D, Stratagouleas ED, Martinez JD. Activation and role of mitogen-activated protein kinases in deoxycholic acid-induced apoptosis. Carcinogenesis. 2001;22:35–41.
  • Yasuda H, Hirata S, Inoue K, et al. Involvement of membrane-type bile acid receptor m-bar/tgr5 in bile acid-induced activation of epidermal growth factor receptor and mitogen-activated protein kinases in gastric carcinoma cells. Biochem Biophys Res Commun. 2007;354:154–159.
  • Goldman A, Chen HD, Roesly HB, et al. Characterization of squamous esophageal cells resistant to bile acids at acidic ph: implication for barrett’s esophagus pathogenesis. Am J Physiol Gastrointest Liver Physiol. 2011;300:G292–G302.
  • Werneburg NW, Yoon JH, Higuchi H, et al. Bile acids activate egf receptor via a tgf-alpha-dependent mechanism in human cholangiocyte cell lines. Am J Physiol Gastrointest Liver Physiol. 2003;285:G31–G36.
  • Rao YP, Stravitz RT, Vlahcevic ZR, et al. Activation of protein kinase c alpha and delta by bile acids: correlation with bile acid structure and diacylglycerol formation. J Lipid Res. 1997;38:2446–2454.
  • Qiao L, Studer E, Leach K, et al. Deoxycholic acid (dca) causes ligand-independent activation of epidermal growth factor receptor (egfr) and fast receptor in primary hepatocytes: inhibition of egfr/mitogen-activated protein kinase-signaling module enhances dca-induced apoptosis. Mol Biol Cell. 2001;12:2629–2645.
  • Looby E, Long A, Kelleher D, et al. Bile acid deoxycholate induces differential subcellular localisation of the pkc isoenzymes beta 1, epsilon and delta in colonic epithelial cells in a sodium butyrate insensitive manner. Int J Cancer. 2005;114:887–895.
  • Keating N, Mroz MS, Scharl MM, et al. Physiological concentrations of bile acids down-regulate agonist induced secretion in colonic epithelial cells. J Cell Mol Med. 2009;13:2293–2303.
  • Fang Y, Han SI, Mitchell C, et al. Bile acids induce mitochondrial ros, which promote activation of receptor tyrosine kinases and signaling pathways in rat hepatocytes. Hepatology. 2004;40:961–971.
  • Jenkins GJS, D’Souza FR, Suzen SH, et al. Deoxycholic acid at neutral and acid ph, is genotoxic to oesophageal cells through the induction of ROS: the potential role of anti-oxidants in Barrett’s oesophagus. Carcinogenesis. 2006;28:136–142.
  • Cronin J, Williams L, McAdam E, et al. The role of secondary bile acids in neoplastic development in the oesophagus. Biochem Soc Trans. 2010;38:337–342.
  • Khare S, Mustafi R, Cerda S, et al. Ursodeoxycholic acid suppresses cox-2 expression in colon cancer: roles of ras, p38, and ccaat/enhancer-binding protein. Nutr Cancer. 2008;60:389–400.
  • Qiao D, Gaitonde SV, Qi W, et al. Deoxycholic acid suppresses p53 by stimulating proteasome-mediated p53 protein degradation. Carcinogenesis. 2001;22:957–964.
  • Song S, Byrd JC, Koo JS, et al. Bile acids induce muc2 overexpression in human colon carcinoma cells. Cancer. 2005;103:1606–1614.
  • Lee HY, Crawley S, Hokari R, et al. Bile acid regulates muc2 transcription in colon cancer cells via positive egfr/pkc/ras/erk/creb, pi3 k/akt/ikappab/nf-kappab and p38/msk1/creb pathways and negative jnk/c-jun/ap-1 pathway. Int J Oncol. 2010;36:941–953.
  • Pyo JS, Ko YS, Kang G, et al. Bile acid induces MUC2 expression and inhibits tumor invasion in gastric carcinomas. J Cancer Res Clin Oncol. 2015;141:1181–1188.
  • Yang HB, Song W, Cheng MD, et al. Deoxycholic acid inhibits the growth of BGC-823 gastric carcinoma cells via a p53‑mediated pathway. Mol Med Rep. 2015;11:2749–2754.
  • Jang ES, Yoon JH, Lee SH, et al. Sodium taurocholate cotransporting polypeptide mediates dual actions of deoxycholic acid in human hepatocellular carcinoma cells: enhanced apoptosis versus growth stimulation. J Cancer Res Clin Oncol. 2014;140:133–144.
  • Brossard D, El Kihel L, Clément M, et al. Synthesis of bile acid derivatives and in vitro cytotoxic activity with pro-apoptotic process on multiple myeloma (KMS-11), glioblastoma multiforme (GBM), and colonic carcinoma (HCT-116) human cell lines. Eur J Med Chem. 2010;45:2912–2918.
  • 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.
  • Huang Y, Cui J, Chen S, et al. Synthesis and antiproliferative activity of C-homo-lactam derivatives of 7-deoxycholic acid. Bioorg Med Chem Lett. 2013;23:2265–2267.
  • Li Q, Liu T, Li Y, et al. Transport and killing mechanism of a novel camptothecin-deoxycholic acid derivate on hepatocellular carcinoma cells. J Drug Target. 2014;22:543–552.
  • Popadyuk II, Markov AV, Salomatina OV, et al. Synthesis and biological activity of novel deoxycholic acid derivatives. Bioorg Med Chem. 2015. Epub 2015 May 13. DOI:10.1016/j.bmc.2015.05.012.
  • Lim SC, Duong HQ, Parajuli KR, et al. Pro-apoptotic role of the MEK/ERK pathway in ursodeoxycholic acid-induced apoptosis in SNU601 gastric cancer cells. Oncol Rep. 2012;28:1429–1434.
  • Shah SA, Volkov Y, Arfin Q, et al. Ursodeoxycholic acid inhibits interleukin beta 1 and deoxycholic acid-induced activation of nf-jb and ap-1 in human colon cancer cells. Int J Cancer. 2006;118:532–539.
  • Peng S, Huo X, Rezaei D, et al. In Barrett’s esophagus patients and Barrett’s cell lines, ursodeoxycholic acid increases antioxidant expression and prevents DNA damage by bile acids. Am J Physiol Gastrointest Liver Physiol. 2014;307:G129–G139.
  • 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.
  • Martínez-Moya P, Romero-Calvo I, Requena P, et al. Dose-dependent antiinflammatory effect of ursodeoxycholic acid in experimental colitis. Int Immunopharmacol. 2013;15:372–380.
  • Hofmann AF, Mysels KJ. Bile acid solubility and precipitation in vitro and in vivo: the role of conjugation, pH, and Ca2+ ions. J Lipid Res. 1992;33:617–626.
  • Hofmann AF, Mysels KJ. Bile salts as biological surfactants. Colloids Surf. 1988;30:145–173.
  • Raimondi F, Santoro P, Barone MV, et al. Bile acids modulate tight junction structure and barrier function of Caco-2 monolayers via EGFR activation. Am J Physiol Gastrointest Liver Physiol. 2008;294:G906–G913.

*A report on mechanism of intestinal membrane permeability enhancement by DOCA

  • Terrell CL, Hughes CE. Antifungal agents used for deep-seated mycotic infections. Mayo Clin Proc. 1992;67:69–91.
  • Baginski M, Czub J. Amphotericin B and its new derivatives – mode of action. Curr Drug Metab. 2009;10:459–469.
  • Torrado JJ, Espada R, Ballesteros MP, et al. Amphotericin B formulations and drug targeting. J Pharm Sci. 2008;97:2405–2425.
  • Guan P, Lu Y, Qi J, et al. Enhanced oral bioavailability of cyclosporine A by liposomes containing a bile salt. Int J Nanomedicine. 2011;6:965–974.
  • Fan D, Wu X, Dong W, et al. Enhancement by sodium caprate and sodium deoxycholate of the gastrointestinal absorption of berberine chloride in rats. Drug Dev Ind Pharm. 2013;39:1447–1456.
  • Sun S, Liang N, Kawashima Y, et al. Hydrophobic ion pairing of an insulin-sodium deoxycholate complex for oral delivery of insulin. Int J Nanomedicine. 2011;6:3049–3056.
  • Park JW, Hwang SR, Jeon OC, et al. Enhanced oral absorption of ibandronate via complex formation with bile acid derivative. J Pharm Sci. 2013;102:341–346.
  • Park JW, Byun Y. Ionic complex of risedronate with positively charged deoxycholic acid derivative: evaluation of physicochemical properties and enhancement of intestinal absorption in rats. Arch Pharm Res. 2014;37:1560–1569.
  • Greenwood J, Adu J, Davey AJ, et al. The effect of bile salts on the permeability and ultrastructure of the perfused, energy-depleted, rat blood-brain barrier. J Cereb Blood Flow Metab. 1991;11:644–654.
  • Yang L, Zhang H, Fawcett JP, et al. Effect of bile salts on the transport of morphine-6-glucuronide in rat brain endothelial cells. J Pharm Sci. 2011;100:1516–1524.
  • Lalić-Popović M, Vasović V, Milijašević B, et al. Deoxycholic acid as a modifier of the permeation of gliclazide through the blood brain barrier of a rat. J Diabetes Res. 2013;2013:598603.
  • Quinn M, McMillin M, Galindo C, et al. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig Liver Dis. 2014;46:527–534.
  • Yu C, Xiao YZ, Xun PH, et al. Preparation and characterization of cucurbitacin B sodium deoxycholate/phospholipid-mixed oral fast dissolving film and antitumor activity study. Zhongguo Zhong Yao Za Zhi. 2014;39:1799–1804.
  • Shi-Xiao J, Shi-Ying J, Lv QY, et al. Mixed nanomicelles loaded with thymopeptides-sodium deoxycholate/phospholipid improve drug absorption. Chin J Nat Med. 2014;12:65–70.
  • Jin S, Fu S, Han J, et al. Improvement of oral bioavailability of glycyrrhizin by sodium deoxycholate/phospholipid-mixed nanomicelles. J Drug Target. 2012;20:615–622.
  • Dong F, Xie Y, Qi J, et al. Bile salt/phospholipid mixed micelle precursor pellets prepared by fluid-bed coating. Int J Nanomedicine. 2013;8:1653–1663.
  • Figueiredo M, Peeke E, DeWitt D, et al. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using same. US20140178475. 2013.
  • Ashford MB, Nolan III JM, Shin E, et al. Therapeutic polymeric nanoparticles and methods of making and using same. US20150140104. 2015.
  • Sakaguchi N. pH sensitive carrier and preparation method thereof, and pH sensitive drug and pH sensitive drug composition each containing the carrier, and method for treating or preventing diseases using the same. US20130323320A1. 2013.
  • Lam KS, Luo J, Lee JS Telodendrimer nanodiscs without apolipoprotein. US8895055. 2014.
  • Lam KS, Li Y, Pan C, et al. Porphyrin modified telodendrimers. US20140161719A1. 2013.
  • Castillo GM, Bolotin EM. Hydrophobic core carrier compositions for delivery of therapeutic agents, methods of making and using the same. US20140154203A1. 2013.
  • Wei W-H, Dong X-M, Liu C-G. In vitro investigation of self-assembled nanoparticles based on hyaluronic acid-deoxycholic acid conjugates for controlled release doxorubicin: effect of degree of substitution of deoxycholic acid. Int J Mol Sci. 2015;16:7195–7209.
  • 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.

*A report on hyaluronic acid-DOCA micelles for delivery of paclitaxel

  • Yan J-K, Ma H-L, 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.

*A report on curdlan-DOCA nanoparticles as a carrier of doxorubicin

  • Mousa SA Composition and method for sulfated non-anticoagulant low molecular weight heparins in cancer and tumor metastasis. US20150132399A1. 2014.
  • Kim S, Choi JW. AIMP2-DX2 gene and its uses. US8431393B2. 2013.
  • Schneider M, Bussat P, Yan F, et al. Assembly of gas-filled microvesicle with active component for contrast imaging. US20110150778A1. 2011.
  • Mousa SA. Nanoformulation of vitamin D derivatives and/or vitamin D metabolites. US8968790B2. 2015.
  • Mousa SA Nanoformulation and methods of use of thyroid receptor beta1 agonists for liver targeting. US20140017329A1. 2013.
  • Mousa SA, Al Haider A, Abdelgader A, et al. Methods and compositions of camel derived products. US20140037722A1. 2013.
  • Mehta NM, Stern W, Gilligan JP. Oral delivery of peptide pharmaceutical compositions. US8835377B2. 2014.
  • Vignati L, DiMarchi RD. Methods for treating metabolic disorders and obesity with a peptide comprising the amino acid sequence of SEQ ID No. 146. US8975223B2. 2015.
  • Byun Y, Lee S, Moon H. Delivery agents for enhancing mucosal absorption of therapeutic agents. US20110237504A1. 2011.
  • Breaker RR, Li S. Antimicrobial compositions and methods. US20150030701A1. 2013.
  • Montagna MT, Lovero G, Coretti C, et al. In vitro activities of amphotericin B deoxycholate and liposomal amphotericin B against 604 clinical yeast isolates. J Med Microbiol. 2014;63(Pt 12):1638–1643.
  • Wa X, McKeon F, Vincent M, et al. Dual function markers for diagnostics and therapeutics for upper gastrointestinal tract precancer. US20150044135A1. 2014.
  • Duncan D, Rubin JP, Golitz L, et al. Refinement of technique in injection lipolysis based on scientific studies and clinical evaluation. Clin Plast Surg. 2009;36:195–209.
  • Young VL. Response to “Injection lipolysis with phosphatidylcholine and deoxycholate”. Aesthet Surg J. 2013;33:1076.
  • Hasengschwandtner F, Gundermann KJ. Injection lipolysis with phosphatidylcholine and deoxycholate. Aesthet Surg J. 2013;33:1071–1072.
  • Salti G, Ghersetich I, Tantussi F, et al. Phosphatidylcholine and sodium deoxycholate in the treatment of localized fat: a double-blind, randomized study. Dermatol Surg. 2008;34:60–66.
  • Duncan D. Commentary on: Metabolic and structural effects of phosphatidylcholine and deoxycholate injections on subcutaneous fat: a randomized, controlled trial. Aesthet Surg J. 2013;33:411–413.
  • Chung SJ, Lee CH, Lee HS, et al. The role of phosphatidylcholine and deoxycholic acid in inflammation. Life Sci. 2014;108:88–93.
  • Lowe NJ. Dissolving unwanted submental fat. Br J Dermatol. 2014;170:237–238.
  • Cook JW, Kennaway EL, Kennaway NM. Production of tumours in mice by deoxycholic acid. Nature. 1940;145:627.
  • Reddy BS, Narasawa T, Weisburger JH, et al. Promoting effect of sodium deoxycholate on colon adenocarcinomas in germfree rats. J Natl Cancer Inst. 1976;56:441–442.
  • LaRue JM, Stratagoules ED, Martinez JD. Deoxycholic acid-induced apoptosis is switched to necrosis by bcl-2 and calphostin C. Cancer Lett. 2000;152:107–113.
  • Barrasa JI, Santiago-Gómez A, Olmo N, et al. Resistance to butyrate impairs bile acid-induced apoptosis in human colon adenocarcinoma cells via up-regulation of Bcl-2 and inactivation of Bax. Biochim Biophys Acta. 2012;1823:2201–2209.
  • Martinez-Augustin O, Sanchez De Medina F. Intestinal bile acid physiology and pathophysiology. World J Gastroenterol. 2008;14:5630–5640.
  • Gangadhar KN, Adhikari K, Srichana T. Synthesis and evaluation of sodium deoxycholate sulfate as a lipid drug carrier to enhance the solubility, stability and safety of an amphotericin B inhalation formulation. Int J Pharm. 2014;471:430–438.

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.