496
Views
35
CrossRef citations to date
0
Altmetric
Reviews

Boron as a platform for new drug design

&
Pages 1017-1027 | Published online: 05 Sep 2012

Bibliography

  • Availablr from http://www.periodic-table.org.uk/element-boron.htm
  • Rezanka T, Sigler K. Biologically active compounds of semi-metals. Phytochem 2008;69:585–606
  • Hawthorne MF. New horizons for therapy based on the boron neutron capture reaction. Mol Med Today 1998;4:174–81
  • Soloway AH, Tjarks W, Barnum BA, The Chemistry of neutron capture therapy. Chem Rev 1998;98:1515–62
  • Armstrong AF, Valliant JF. The bioinorganic and medicinal chemistry of carboranes: from new drug discovery to molecular imaging and therapy. Dalton Trans 2007;38:4240–51
  • Barth RF, Coderre JA, Vicente MG, Boron neutron capture therapy of cancer: current status and future prospects. Clin Cancer Res 2005;11:3987–4002
  • Baker SJ, Ding CZ, Akama T, Therapeutic potential of boron-containing compounds. Future Med Chem 2009;1:1275–88
  • Fujii S, Masuno H, Taoda Y. Boron cluster-based development of potent nonsecosteroidal vitamin d receptor ligands: direct observation of hydrophobic interaction between protein surface and carborane. JACS 2011;133:20933–41
  • Calvaresi M, Zerbetto F. In silico carborane docking to proteins and potential drug targets. J Chem Inf Model 2011;51:1882–96
  • Fischli W, Leukart O, Schwyzer R. Hormone-Receptor interactions. carboranylalanine (car) as a phenylalanine analogue: reactions with chymotrypsin. Helv Chim Acta 1977;60:959–63
  • Leukart O, Escher E, Schwyzer R. Synthesis of angiotensins, bradykinins and substance P octapeptides in which the residues Phe and Tyr have been replaced with car and of [Car1, Leu5]-enkephalin. Helv Chim Acta 1979;62:546–52
  • Fauchere JL, Leukart O, Eberle A, Schwyzer R. The synthesis of [4-Carboranylalanine, 5-Leucine]-Enkephalin (Including an Improved Preparation of t-Butoxycarbonyl-L-o-carboranylalnine, New Derivatives of L-Propargylglycine, and a Note on Melanotropic and Opiate Receptor Binding Characteristics). Helv Chim Acta 1979;62:1385–95
  • Issa F, Kassiou M, Rendina LM. Boron in drug discovery: carboranes as unique pharmacophores in biologically active compounds. Chem Rev 2011;111:5701–22
  • Sivaev IB, Bregadze VV. Polyhedral boranes for medical applications: current status and perspectives. Eur J Inorg Chem 2009;11:1433–50
  • Lesnikowski ZJ. Boron units as pharmacophores – new applications and opportunities of boron cluster chemistry. Collect Czech Chem Commun 2007;72:1646–58
  • Golberg D, Bando Y, Tang C, Zhi C. Boron nitride nanotubes. Adv Mater 2007;19:2413–32
  • Terrones M, Romo-Herrera JM, Cruz-Silva E, Pure and doped boron nitride nanotubes. Mater Today 2007;10:30–8
  • Lacerda L, Raffa V, Prato M, Cell-penetrating CNTs for delivery of therapeutics. Nano Today 2007;2:38–43
  • Suryavanshi AP, Yu MF, Wen J, Elastic modulus and resonance behavior of boron nitride nanotubes. Appl Phys Lett 2004;84:2527–9
  • Chen Y, Zou J, Campbell SJ, Le Caer G. Boron nitride nanotubes: pronounced resistance to oxidation, Appl Phys Lett. 2004;84:2430–2
  • Blasé X, Rubio A, Louie SG, Cohen ML. Quasiparticle band structure of bulk hexagonal boron nitride and related systems. Phys Rev B 1995;51:6868–75
  • Ciofani G, Raffa V, Meniassi A, Cuschieri A. Cytocompatibility, Interactions, and Uptake of polyethyleneimine-coated boron nitride nanotubes by living cells: confirmation of their potential for biomedical applications. Biotech Bioeng 2008;101:850–8
  • Ciofani G, Gerchi GG, Liakos I, A simple approach to covalent functionalization of boron nitride nanotubes. J Coll Interf Sci 2012;374:308–14
  • Pantarotto D, Briand JP, Prato M, Bianco A. Translocation of bioactive peptides across cell membranes by carbon nanotubes. Chem Commun 2004;1:16–117
  • Shi Kam N, Liu Z, Dai H. Carbon nanotubes as intracellular transporters for proteins and DNA: an investigation of the uptake mechanism and pathway. Angew Chem Int 2006;45:577–81
  • Kostarelos K, Lacerda L, Pastorin G, Cellular uptake of functionalized carbon nanotubes is independent of functional group and cell type. Nat Nanotechnol 2007;2:108–13
  • Bai X, Golberg D, Bando Y, Deformation-driven electrical transport of individual boron nitride nanotubes. Nano Lett 2007;7:632–7
  • Ciofani G, Danti S, D'Alessandro D, Enhancement of neurite outgrowth in neuronal-like cells following boron nitride nanotube-mediated stimulation. ACSNano 2010;4:6267–77
  • Della Flora E, Perera CL, Cameron AL, Maddern GJ. Deep brain stimulation for essential tremor: a systematic review. Movement Disord 2010;25:1550–9
  • Xu J, Chen JDZ. Intestinal electrical stimulation improves delayed gastric emptying and vomiting induced by duodenal distension in dogs. Neurogastroenterol Motil 2008;20:236–42
  • Ross KB, Dubin S, Nigroni P, Programmed stimulation for simulation of atrial tachyarrythmias. Biomed Sci Instrum 1997;33:25–9
  • Gordon T, Brushart TM, Amirjani N, Chan KM. The potential of electrical stimulation to promote functional recovery after peripheral nerve injury - comparisons between rats and humans. Acta Neurochir 2007;100:3–11
  • Halavaara J, Tervahartiala P, Isonieme H, Hockerstedt K. Efficacy of sequential use of supeparamagnetic iron oxide and gadolinium in liver MR imaging. Acta Radiol 2002;43:180–5
  • Hilger I, Fruhauf K, Andra W, Heating potential of iron oxides for therapeutic purposes in interventional radiology. Acad Radiol 2002;9:198–202
  • Luebbe AS, Alexiou C, Bergemann C. Clinical applications of magnetic drug targeting. J Surg Res 2001;95:200–6
  • Ciofani G, Raffa V, Obata Y, Magnetic driven alginate nanoparticles for targeted drug delivery. Curr Nanosci 2008;4:212–18
  • Arruebo M, Fernandez-Pacheco R, Ibarra MR, Santamaria J. Magnetic nanoparticles for drug delivery. Nano Today 2007;2:22–32
  • Ciofani G, Raffa V, Yu J, Boron nitride nanotubes: a novel vector for targeted magnetic drug delivery. Curr Nanosci 2009;5:33–8
  • Cheng F, Jakle F. Boron-containing polymers as versatile building blocks for functional nanostructured materials. Polym Chem 2011;2:2122–32
  • Jakle F. Advances in the synthesis of organoborane polymers for optical, electronic, and sensory applications. Chem Rev 2010;110:3985–4022
  • Entwistle CD, Marder TB. Applications of three-coordinate organoboron compounds and polymers in optoelectronics. Chem Mater 2004;16:4574–85
  • Zhang G, Chen J, Payne SJ, Multi-Emissive difluoroboron dibenzoylmethane polylactide exhibiting intense fluorescence and oxygen-sensitive room-temperature phosphorescence. JACS 2007;129:8942–3
  • Pfister A, Zhang G, Zareno J, Horwitz AF Fraser CL. Boron Polylactide nanoparticles exhibiting fluorescence and phosphorescence in aqueous medium. ACS Nano 2008;2:1252–1258
  • Zhang G, Palmer GM, Dewhirst MW, Fraser CL. A dual-emissive-materials design concept enables tumour hypoxia imaging. Nat Mater 2009;9:747–51
  • Kersey FR, Zhang GQ, Palmer GM, Stereocomplexed poly(lactic acid)−Poly(ethylene glycol) nanoparticles with dual-emissive boron dyes for tumor accumulation. ACS Nano 2010;4:4989–96
  • Ulrich G, Ziessel R, Harriman A. The chemistry of fluorescent BODIPY dyes: versatility unsurpassed. Angew Chem Int Ed 2008;47:1184–201
  • Ahmad AA, Alsaad AM. Adhesive B-doped DLC films on biomedical alloys used for bone fixation. Bull Mater Sci 2007;30:301–8
  • Li DJ, Gu HQ. Cell attachment on diamond-like carbon coating. Bull Mater Sci 2002;25:7–13
  • Hauert R. A review of modified DLC coatings for biological applications. Dia Relat Mater 2003;12:583–9
  • Lee HJ, Lee JK, Zubeck R, Properties of sputter-deposited hydrogenated carbon films as a tribological overcoat used in rigid magnetic disks. Surf Coating Technol 1992;54:55:552–6
  • Miyoshi K, Wu RL, Garscadden A. Friction and wear of diamond and diamond-like carbon coatings. Surf Coat Technol 1992;54:55:428–34
  • Harris SJ, Weiner AM, Tung SC, A diamond-like carbon film for wear protection of steel. Surf Coating Technol 1993;62:550–7
  • Hench LL, Wilson J. Surface-active biomaterials. Science 1984;226:630–6
  • Silver IA, Deas 00, Erecińska M. Interactions of bioactive glasses with osteoblasts in vitro: effects of 45S5 Bioglass, and 58S and 77S bioactive glasses on metabolism, intracellular ion concentrations and cell viability. Biomaterials 2001;22:175–85
  • Chen QZ, Thompson ID, Boccaccini AR. 45S5 Bioglassw-derived glass–ceramic scaffolds for bone tissue engineering. Biomaterials 2006;27:2414–25
  • Hamadouche M, Meunier A, Greenspan DC, Long-term in vivo bioactivity and degradability of bulk sol-gel bioactive glasses. J Biomed Mater Res 2001;54:560–6
  • Pan HB, Zhao XL, Zhang X, Strontium borate glass: potential biomaterial for bone regeneration. J R Soc Interface 2010;7:1025–31
  • Ellis GA, Palte MJ, Raines RT. Boronate-Mediated biologic delivery. JACS 2012;134:3631-3634
  • Varki A, Cummings RD, Esko JD, Essentials of glycobiology. 2nd edition. Cold Spring Harbor Laboratory Press; Cold Spring Harbor NY: 2009
  • James TD, Phillips MD, Shinkai S. Boronic acids in saccharide recognition. Royal Society of Chemistry; Cambridge, UK: 2006
  • Zhong X, Bai HJ, Xu JJ, Reusable interface constructed by 3-aminophenylboronic acid-functionalized multiwalled carbon nanotubes for cell capture, release, and cytosensing. Adv Funct Mater 2010;20:992–9
  • Matsumoto A, Cabral H, Sato N, Assessment of tumor metastasis by the direct determination of cell-membrane sialic acid expression. Angew Chem Int Ed 2010;49:5494–7
  • Wu W, Mitra N, Yan EC, Zhou S. Multifunctional hybrid nanogel for integration of optical glucose sensing and self-regulated insulin release at physiological pH. ACS Nano 2010;4:4831–9
  • Kumar A, Hozo I, Wheatley K, Djulbegovic B. Thalidomide versus bortezomib based regimens as first-line therapy for patients with multiple myeloma: a systematic review. Am J Hematol 2011;86:18–24
  • Peng Q, Chen F, Zhong Z, Zhuo R. Enhanced gene transfection capability of polyethylenimine by incorporating boronic acid groups. Chem Commun 2010;46:5888–90
  • Piest M. Engbersen JFJ. Role of boronic acid moieties in poly(amido amine)s for gene delivery. J Control Release 2011;155:331–40
  • Soloway AH, Hatanaka H, Davis MA. Penetration of brain and brain tumor. VII. Tumor-binding sulfhydryl boron compounds. J Med Chem 1967;10:714–17
  • Nakagawa Y, Hatanaka H. Boron neutron capture therapy: clinical brain tumor studies. J Neuro Oncol 1997;33:105–15
  • Snyder HR, Reedy AJ, Lennarz WJ. Synthesis of aromatic boronic acids. aldehydo boronic acids and a boronic acid analog of tyrosine. JACS 1958;80:835–8
  • Mishima Y, Ichihashi M, Hatta S, New thermal neutron capture therapy for malignant melanoma: melanogenesis-seeking 10B molecule-melanoma cell interaction from in vitro to first clinical trial. Pigment Cell Res 1989;2:226–34
  • Yanagie H, Ogata A, Suguyama H, Application of drug delivery system to boron neutron capture therapy for cancer. Exp Opin Drug Deliv 2008;5:427–43
  • Barth RF, Yang W, Al-Madhoun AS, Boron-containing nucleosides as potential delivery agents for neutron capture therapy of brain tumors. Cancer Res 2004;64:6287–95
  • Lesnikowski ZJ, Shi J, Schinazi RF. Nucleic acids and nucleosides containing carboranes. J Oganometallic Chem 1999;581:156–69
  • Matejícek P, Cígler P, Olejniczak AB, Aggregation behavior of nucleoside-boron cluster conjugates in aqueous solutions. Langmuir 2008;24:2625–30
  • Olejniczak AB, Semenuk A, Kwiatkowski M, Lesnikowski ZJ. Synthesis of adenosine containing carborane modification. J Oganometallic Chem 2003;680:124–6
  • Wojtczak B, Semenyuk A, Olejniczak AB, General method for the synthesis of 2′-O-carboranyl-nucleosides. Tetrahedron Lett 2005;46:3969–72
  • Kabalka GV, Yao ML. Synthesis of a novel boronated 1-aminocyclobutanecarboxylic acid as a potential boron neutron capture therapy agent. App Organomet Chem 2003;17:398–402
  • Manusaga SI, Ono K, Kirihata M, Potential of a-amino alcohol p-boronophenylalaninol as a boron carrier in boron neutron capture therapy, regarding its enantiomers. J Canc Res Clin Oncol 2003;129:21–8
  • Tietze LF, Bothe U. Ortho-carboranyl glycosides of glucose, mannose, maltose and lactose for cancer treatment by boron neutron-capture therapy. Chem Eur J 1998;4:1179–83
  • Giovenzana GB, Lay L, Monti D, Synthesis of carboranyl derivatives of alkynyl glycosides as potential BNCT agents. Tetrahedron 1999;55:14123–36
  • Tietze LF, Griesbach U, Schuberth I, Novel carboranyl C-glycosides for the treatment of cancer by boron neutron capture therapy. Chem An Eur J 2003;9:1296–1302
  • Orlova AV, Kononov LO, Kimel BG, Conjugates of polyhedral boron compounds with carbohydrates. 4. Hydrolytic stability of carborane–lactose conjugates depends on the structure of a spacer between the carborane cage and sugar moiety. Appl Organometal Chem 2006;20:416–20
  • Lee JD, Ueno M, Miyajima Y, Nakamura H. Synthesis of boron cluster lipids: closo-dodecaborate as an alternative hydrophilic function of boronated liposomes for neutron capture therapy. Oganic Lett 2007;9:323–6
  • Fabris C, Jori G, Giuntini F, Roncucci G. Photosensitizing properties of a boronated phthalocyanine: studies at the molecular and cellular level. J Photochem Photobiol B 2001;64:1–7
  • Vicente MGH, Wickramasinghe A, Nurco DJ. Synthesis, toxicity and biodistribution of two 5,15-Di[3,5-(nidocarboranylmethyl) phenyl]porphyrins in EMT-6 tumor bearing mice. Bioorg Med Chem 2003;11:3101–8
  • Friso E, Roncucci G, Dei D, A novel 10B-enriched carboranyl-containing phthalocyanine as a radio- and photo-sensitising agent for boron neutron capture therapy and photodynamic therapy of tumours: in vitro and in vivo studies. Photochem Photobiol Sci 2006;5:39–50
  • Ristori S, Salvati A, Martini G, Synthesis and liposome insertion of a new poly(carboranylalkylthio)porphyrazine to improve potentiality in multiple-approach cancer therapy. JACS 2007;129:2728–9
  • Salvati A, Ristori S, Obersisse J, Small angle scattering and zeta potential of liposomes loaded with octa(carboranyl)porphyrazine. J Phys Chem B 2007;111:10357–64
  • Jori G, Soncin M, Friso E, A novel boronated-porphyrin as a radio-sensitizing agent for boron neutron capture therapy of tumors: in vitro and in vivo studies. Appl Rad Isot 2009;67:S321–4
  • Renner MW, Miura M, Easson MW, Recent progress in the syntheses and biological evaluation of boronated porphyrins for boron neutron-capture therapy anti-cancer agents. Med Chem 2006;6:6145–57
  • Yang W, Barth RF, Wu G, Molecular targeting and treatment of EGFRvIII-positive gliomas using boronated monoclonal antibody L8A4. Clin Cancer Res 2006;12:3792–802
  • Yang W, Barth RF, Wu G, Boron neutron capture therapy of EGFR or EGFRvIII positive gliomas using either boronated monoclonal antibodies or epidermal growth factor as molecular targeting agents. Appl Rad Isot 2009;67:S328–31
  • Hawthorne MF, Shelli K. Liposomes as drug delivery vehicles for boron agents. J Neuro Oncol 1997;33:53–8
  • Ristori S, Oberdisse J, Grillo I, Structural characterization of cationic liposomes loaded with sugar-based carboranes. Biophys J 2005;88:535–47
  • Li T, Hamdi J, Hawthorne MF. Unilamellar liposomes with enhanced boron content. Bioconj Chem 2006;17:15–20
  • Altieri S, Balzi M, Bortolussi S, Carborane derivatives loaded into liposomes as efficient delivery systems for boron neutron capture therapy. J Med Chem 2009;52:7829–35
  • Ma L, Hamdi F, Huang J, Hawthorne MF. Camouflaged carborane amphiphiles:  synthesis and self-assembly. Inorg Chem 2005;44:7249–58
  • Ma L, Hamdi F, Wong F, Hawthorne MF. Closomers of high boron content: synthesis, characterization, and potential application as unimolecular nanoparticle delivery vehicles for boron neutron capture therapy. Inorg Chem 2006;45:278–85
  • Galie KM, Mollard A, Zharov I. Polyester-based carborane-containing dendrons. Inorg Chem 2006;45:7815–20
  • Parrott MC, Marchington EB, Valliant JF, Adronov A. Synthesis and Properties of Carborane-Functionalized Aliphatic Polyester Dendrimers. JACS 2005;127:12081–9
  • Petersen MS, Petersen CC. Agger boron nanoparticles inhibit tumour growth by boron neutron capture therapy in the murine B16-OVA model. Anticancer Res 2008;28:571–6
  • Baše T, Bastl Z, Slouf M, Gold micrometer crystals modified with carboranethiol derivatives. J Phys Chem 2008;112:14446–55
  • Zhu Y, Lin Y, Zhu YZ. Boron drug delivery via encapsulated magnetic nanocomposites: a new approach for BNCT in cancer treatment. J Nanomat 2010;2010:8 ID 409320
  • Mandal S, Bakeine GJ, Krol S, Design, development and characterization of multifunctionalized gold nanoparticles for biodetection and targeted Boron delivery in BNCT applications. Appl Radiat Isot 2011;69:1692–7
  • Cheng F, Jäkle F. Boron-containing polymers as versatile building blocks for functional nanostructured materials. Polym Chem 2011;2:2122–32
  • Sumitani S, Yukio N. Boron neutron capture therapy assisted by boron-conjugated nanoparticles. Polymer J 2012;44:522–30
  • Yanagie H, Fujii Y, Takahashi T, Boron neutron capture therapy using 10B entrapped anti-CEA immunoliposome. Hum Cell 1989;2:290–6
  • Yanagie H, Tomita T, Kobayashi H, Application of boronated anti-CEA immunoliposome to tumour cell growth inhibition in in vitro boron neutron capture therapy model. Br J Cancer 1991;63:522–6
  • Pan X, Wu G, Yang W, Synthesis of cetuximab-immunoliposomes via a cholesterol-based membrane anchor for targeted delivery of a Neutron Capture Therapy (NCT) agent to glioma cells. Bioconj Chem 2007;18:101–8
  • Pan XQ, Wang H, Shukla S, Boron-containing folate receptor-targeted liposomes as potential delivery agents for neutron capture therapy. Bioconj Chem 2002;13:435–42
  • Kullberg EB, Carlsson J, Edwards K. Introductory experiments on ligand liposomes as delivery agents for boron neutron capture therapy. Int JOncol 2003;23:461–7
  • Maruyama K, Ishida O, Kasaoka S, Intracellular targeting of sodium mercaptoundecahydrododecaborate (BSH) to solid tumors by transferrin-PEG liposomes, for boron neutron-capture therapy (BNCT). J Control Release 2004;98:195–335
  • Yanagie H, Ogura K, Takagi K. Accumulation of boron compounds to tumor with polyethylene-glycol binding liposome by using neutron capture autoradiography. Appl Radiat Isot 2004;61:639–46
  • Barth RF, Yanga W, Wua G. Thymidine kinase 1 as a molecular target for boron neutron capture therapy of brain tumors. PNAS 2008;105:17493–7
  • Kroto HW, Heath JR, O'Brien SC, C60: buckminsterfullerene. Nature 1985;318:162–3
  • Green TA, Switendick AC, Emin D. Ab Initio Self-Consistent Field (SCF) calculations on borane icosahedra with zero, one, or two substituted carbon atoms. J Chem Phys 1988;89:6815–22
  • Schleyer PVR, Najafian K. Stability and three-dimensional aromaticity of closo-monocarbaborane anions, CBn-1Hn-,and closo-dicarboranes, C2Bn-2Hn. Inorg Chem 1998;37:3454–70
  • Oliva JM, Klein DJ, von Ragué-Schleyer P, Serrano-Andrés L. Design of carborane molecular architectures with electronic structure computations: from endohedral and polyradical systems to multidimensional networks. Pure appl Chem 2009;81:719–29

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.