438
Views
2
CrossRef citations to date
0
Altmetric
Research Articles

Design, synthesis, spectroscopic characterizations, single crystal X-ray analysis, in vitro xanthine oxidase and acetylcholinesterase inhibitory evaluation as well as in silico evaluation of selenium-based N-heterocyclic carbene compounds

, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 11728-11747 | Received 08 Jul 2022, Accepted 22 Dec 2022, Published online: 09 Jan 2023

References

  • Accelrys Software Inc. (2013). Discovery studio modeling environment. Release 3.5 Accelrys Software Inc.
  • Aktaş, A., Celepci, D. B., Gök, Y., & Aygün, M. (2018). 2‐hydroxyethyl‐substituted (NHC) Pd(II)PPh3 complexes: Synthesis, characterization, crystal structure and ıts application on sonogashira cross‐coupling reactions in aqueous media. ChemistrySelect, 3(39), 10932–10937. https://doi.org/10.1002/slct.201802519
  • Aktaş, A., Noma, S. A. A., Celepci, D. B., Erdemir, F., Gök, Y., & Ateş, B. (2019). New 2-hydroxyethyl substituted N-heterocyclic carbene precursors: Synthesis, characterization, crystal structure and inhibitory properties against carbonic anhydrase and xanthine oxidase. Journal of Molecular Structure, 1184, 487–494. https://doi.org/10.1016/j.molstruc.2019.02.063
  • Aldrich, P. E., & Sheppard, W. A. (1964). α-Fluorinated ethers. II. Alkyl fluoroalkyl ethers1. The Journal of Organic Chemistry, 29(1), 11–15. https://doi.org/10.1021/jo01024a002
  • Arduengo, A. J., III, Harlow, R. L., & Kline, M. (1991). A stable crystalline carbene. Journal of the American Chemical Society, 113(1), 361–363. https://doi.org/10.1021/ja00001a054
  • Bejoymohandas, K. S., Kumar, A., Varughese, S., Varathan, E., Subramanian, V., & Reddy, M. L. P. (2015). Photophysical and electroluminescence properties of bis (2′, 6′-difluoro-2, 3′-bipyridinato-N, C 4′) iridium (picolinate) complexes: Effect of electron-withdrawing and electron-donating group substituents at the 4′ position of the pyridyl moiety of the cyclometalated ligand. Journal of Materials Chemistry C, 3(28), 7405–7420. https://doi.org/10.1039/C5TC01260K
  • Bhowmick, S., Saha, A., Osman, S. M., Alasmary, F. A., Almutairi, T. M., & Islam, M. A. (2021). Structure-based identification of SARS-CoV-2 main protease inhibitors from anti-viral specific chemical libraries: An exhaustive computational screening approach. Molecular Diversity, 25(3), 1979–1997. https://doi.org/10.1007/s11030-021-10214-6
  • Blumberg, D., Doron, S., & Bitton, S. (1985). Effect of triflumuron on two species of nitidulid beetles, Carpophilus hemipterus and Urophorus humeralis. Phytoparasitica, 13(1), 9–19. https://doi.org/10.1007/BF02994433
  • Boban, M., Kocic, G., Radenkovic, S., Pavlovic, R., Cvetkovic, T., Deljanin-Ilic, M., Ilic, S., Bobana, M. D., Djindjic, B., Stojanovic, D., Sokolovic, D., & Jevtovic-Stoimenov, T. (2014). Circulating purine compounds, uric acid, and xanthine oxidase/dehydrogenase relationship in essential hypertension and end stage renal disease. Renal Failure, 36(4), 613–618. https://doi.org/10.3109/0886022X.2014.882240
  • Bockfeld, D., Bannenberg, T., Jones, P. G., & Tamm, M. (2017). N‐heterocyclic carbene adducts of phenyldioxophosphorane and its heavier sulfur and selenium analogues. European Journal of Inorganic Chemistry, 2017(28), 3452–3458. https://doi.org/10.1002/ejic.201700494
  • Burmaoglu, S., Ozcan, S., Balcioglu, S., Gencel, M., Noma, S. A. A., Essiz, S., Ates, B., & Algul, O. (2019). Synthesis, biological evaluation and molecular docking studies of bis-chalcone derivatives as xanthine oxidase inhibitors and anticancer agents. Bioorganic Chemistry, 91, 103149. https://doi.org/10.1016/j.bioorg.2019.103149
  • Bursal, E., Taslimi, P., Gören, A. C., & Gülçin, İ. (2020). Assessments of anticholinergic, antidiabetic, antioxidant activities and phenolic content of Stachys annua. Biocatalysis and Agricultural Biotechnology, 28, 101711. https://doi.org/10.1016/j.bcab.2020.101711
  • Bytyqi-Damoni, A., Genç, H., Zengin, M., Beyaztas, S., Gençer, N., & Arslan, O. (2012). In vitro effect of novel β-lactam compounds on xanthine oxidase enzyme activity. Artificial Cells, Blood Substitutes, and İmmobilization Biotechnology, 40(6), 369–377. https://doi.org/10.3109/10731199.2012.678943
  • Chen, Y., Zhang, A. F., Wang, W. X., Zhang, Y., & Gao, T. C. (2012). Baseline sensitivity and efficacy of thifluzamide in Rhizoctonia solani. Annals of Applied Biology, 161(3), 247–254. https://doi.org/10.1111/j.1744-7348.2012.00569.x
  • Clark, R. C., & Reid, J. S. (1995). The analytical calculation of absorption in multifaceted crystals. Acta Crystallographica Section A Foundations of Crystallography, 51(6), 887–897. https://doi.org/10.1107/S0108767395007367
  • CrysAlisPro Software. (2020). Version 1.171.41.93a. Rigaku Corporation.
  • DrugBank. (n.d.). Retrieved June 8, 2022, from www.drugbank.ca
  • Dolomanov, O. V., Bourhis, L. J., Gildea, R. J., Howard, J., A. K., & Puschmann, H. (2009). OLEX2: A complete structure solution, refinement and analysis program. Journal of Applied Crystallography, 42(2), 339–341. https://doi.org/10.1107/S0021889808042726
  • Dumas, J. B., & Peligot, E. M. (1835). Mémoire sur l‘esprit de bois et sur les divers composés éthérés qui en proviennent. Annales de Chimie et de Physique, 58, 5–74.
  • Ellman, G. L., Courtney, K. D., Andres Jr, V., & Featherstone, R. M. (1961). A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol, 7(2), 88–95.
  • Erdemir, F., Aktaş, A., Barut Celepci, D., & Gök, Y. (2020). New (NHC) Pd (II)(PPh3) complexes: Synthesis, characterization, crystal structure and its application on Sonogashira and Mizoroki–Heck cross-coupling reactions. Chemical Papers, 74(1), 99–112. https://doi.org/10.1007/s11696-019-00859-x
  • Ezugwu, C. I., Kabir, N. A., Yusubov, M., & Verpoort, F. (2016). Metal–organic frameworks containing N-heterocyclic carbenes and their precursors. Coordination Chemistry Reviews, 307, 188–210. https://doi.org/10.1016/j.ccr.2015.06.012
  • Fernandes, A. P., & Gandin, V. (2015). Selenium compounds as therapeutic agents in cancer. Biochimica Et Biophysica Acta, 1850(8), 1642–1660. https://doi.org/10.1016/j.bbagen.2014.10.008
  • Frisch, M. J., Trucks, G. W., Schlegel, H. B., Scuseria, G. E., Robb, M. A., Cheeseman, J. R., Scalmani, G., Barone, V., Mennucci, B., & Petersson, G. A. (2009). Gaussian 09, Revision E.01. Gaussian, Inc.
  • Gillis, E. P., Eastman, K. J., Hill, M. D., Donnelly, D. J., & Meanwell, N. A. (2015). Applications of fluorine in medicinal chemistry. Journal of Medicinal Chemistry, 58(21), 8315–8359. https://doi.org/10.1021/acs.jmedchem.5b00258
  • GNU General Public License. (1989). Retrieved December 25, 2014.
  • Grygorenko, O. O., Volochnyuk, D. M., Ryabukhin, S. V., & Judd, D. B. (2020). The symbiotic relationship between drug discovery and organic chemistry. Chemistry (Weinheim an Der Bergstrasse, Germany), 26(6), 1196–1237. https://doi.org/10.1002/chem.201903232
  • Gu, C., Howell, K., Dunshea, F. R., & Suleria, H. A. (2019). Lc-esi-qtof/ms characterisation of phenolic acids and flavonoids in polyphenol-rich fruits and vegetables and their potential antioxidant activities. Antioxidants, 8(9), 405. https://doi.org/10.3390/antiox8090405
  • Halliwell, B., Gutteridge, J. C., & Cross, C. E. (1992). Free radicals, antioxidants, and human disease: Where are we now? The Journal of Laboratory and Clinical Medicine, 119(6), 598–620.
  • Han, J., Remete, A. M., Dobson, L. S., Kiss, L., Izawa, K., Moriwaki, H., Soloshonok, V. A., & O’Hagan, D. (2020). Next generation organofluorine containing blockbuster drugs. Journal of Fluorine Chemistry, 239, 109639. https://doi.org/10.1016/j.jfluchem.2020.109639
  • Hayat, K., Tariq, U., Wong, Q. A., Quah, C. K., Majid, A. S. A., Nazari V. M., Ahamed, M. B. K., Iqbal, M. A., & Tirmizi, S. A. (2021). Green synthesis of selenium based N-heterocyclic carbene compounds; structural, in-vitro anticancer and molecular docking studies. Computational Biology and Chemistry, 94, 107567.
  • Hoehn, R. D., Carignano, M. A., Kais, S., Zhu, C., Zhong, J., Zeng, X. C., Francisco, J. S., & Gladich, I. (2016). Hydrogen bonding and orientation effects on the accommodation of methylamine at the air-water interface. The Journal of Chemical Physics, 144(21), 214701.
  • Hopkinson, M. N., Richter, C., Schedler, M., & Glorius, F. (2014). An overview of N-heterocyclic carbenes. Nature, 510(7506), 485–496. https://doi.org/10.1038/nature13384
  • Huang, J., Grasa, G., & Nolan, S. P. (1999). General and efficient catalytic amination of aryl chlorides using a palladium/bulky nucleophilic carbene system. Organic Letters, 1(8), 1307–1309. https://doi.org/10.1021/ol990987d
  • Iqbal, M. A., Haque, R. A., Ng, W. C., Hassan, L. E., Majid, A. M., & Razali, M. R. (2016). Green synthesis of mono-and di-selenium-N-heterocyclic carbene adducts: Characterizations, crystal structures and pro-apoptotic activities against human colorectal cancer. Journal of Organometallic Chemistry, 801, 130–138. https://doi.org/10.1016/j.jorganchem.2015.10.023
  • Iqbal, N., Yaqoob, M., Javed, M., Abbasi, M., Iqbal, J., & Iqbal, M. A. (2021). Synthesis in combination with biological and computational evaluations of selenium-N-heterocyclic carbene compounds. Computational and Theoretical Chemistry, 1197, 113135. https://doi.org/10.1016/j.comptc.2020.113135
  • Jamil, M. S. S., & Endot, N. A. (2020). Influence of fluorine substituents on the electronic properties of selenium-N-heterocyclic carbene compounds. Molecules, 25(21), 5161. https://doi.org/10.3390/molecules25215161
  • Jo, S., Kim, T., Iyer, V. G., & Im, W. (2008). CHARMM‐GUI: A web‐based graphical user interface for CHARMM. Journal of Computational Chemistry, 29(11), 1859–1865. https://doi.org/10.1002/jcc.20945
  • Kamal, A., Iqbal, M. A., Bhatti, H. N., & Ghaffar, A. (2022). Selenium-N-heterocyclic carbene (Se-NHC) complexes with higher aromaticity inhibit microbes: Synthesis, structure, and biological potential. Journal of Coordination Chemistry, 75(11-14), 1915–1928.
  • Kamal, A., Nazari, M., Yaseen, M., Iqbal, M. A., Ahamed, M. B. K., Majid, A. S. A., & Bhatti, H. N. (2019). Green synthesis of selenium-N-heterocyclic carbene compounds: Evaluation of antimicrobial and anticancer potential. Bioorganic Chemistry, 90, 103042.
  • Kondratiuk, V. Y., Tarasenko, O. M., & Synytsya, Y. P. (2016). Peculiarity of nutrition in patients with gout. Scientific Reviews, 1(44).
  • Meanwell, N. A. (2011). Improving drug candidates by design: A focus on physicochemical properties as a means of improving compound disposition and safety. Chemical Research in Toxicology, 24(9), 1420–1456. https://doi.org/10.1021/tx200211v
  • Meanwell, N. A. (2018). Fluorine and fluorinated motifs in the design and application of bioisosteres for drug design. Journal of Medicinal Chemistry, 61(14), 5822–5880. https://doi.org/10.1021/acs.jmedchem.7b01788
  • Mei, H., Remete, A. M., Zou, Y., Moriwaki, H., Fustero, S., Kiss, L., Soloshonok, V. A., & Han, J. (2020). Fluorine-containing drugs approved by the FDA in 2019. Chinese Chemical Letters, 31(9), 2401–2413. https://doi.org/10.1016/j.cclet.2020.03.050
  • Mei, H., Han, J., Fustero, S., Medio-Simon, M., Sedgwick, D. M., Santi, C., Ruzziconi, R., & Soloshonok, V. A. (2019). Fluorine‐containing drugs approved by the FDA in 2018. Chemistry (Weinheim an Der Bergstrasse, Germany), 25(51), 11797–11819. https://doi.org/10.1002/chem.201901840
  • Misra, S., Boylan, M., Selvam, A., Spallholz, J. E., & Björnstedt, M. (2015). Redox-active selenium compounds—From toxicity and cell death to cancer treatment. Nutrients, 7(5), 3536–3556. https://doi.org/10.3390/nu7053536
  • Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., Goodsell, D. S., & Olson, A. J. (2009). AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry, 30(16), 2785–2791. https://doi.org/10.1002/jcc.21256
  • Nair, V., Bindu, S., & Sreekumar, V. (2004). N‐heterocyclic carbenes: Reagents, not just ligands!. Angewandte Chemie International Edition, 43(39), 5130–5135. https://doi.org/10.1002/anie.200301714
  • Nakamura, T., Murase, T., Nampei, M., Morimoto, N., Ashizawa, N., Iwanaga, T., & Sakamoto, R. (2016). Effects of topiroxostat and febuxostat on urinary albumin excretion and plasma xanthine oxidoreductase activity in db/db mice. European Journal of Pharmacology, 780, 224–231. https://doi.org/10.1016/j.ejphar.2016.03.055
  • Nile, S. H., Kumar, B., & Park, S. W. (2013). In vitro evaluation of selected benzimidazole derivatives as an antioxidant and xanthine oxidase inhibitors. Chemical Biology & Drug Design, 82(3), 290–295. https://doi.org/10.1111/cbdd.12141
  • Öfele, K. (1968). 1, 3-Dimethyl-4-imidazolinyliden-(2)-pentacarbonylchrom ein neuer Übergangsmetall-carben-komplex. Journal of Organometallic Chemistry, 12(3), P42–P43. https://doi.org/10.1016/S0022-328X(00)88691-X
  • Onul, N., Ertik, O., Mermer, N., & Yanardag, R. (2018). Synthesis and biological evaluation of S-substituted perhalo-2-nitrobuta-1, 3-dienes as novel xanthine oxidase, tyrosinase, elastase, and neuraminidase inhibitors. Journal of Chemistry, 2018, 1–11. https://doi.org/10.1155/2018/4386031
  • Pacher, P. A. L., Nivorozhkin, A., & Szabó, C. (2006). Therapeutic effects of xanthine oxidase inhibitors: Renaissance half a century after the discovery of allopurinol. Pharmacological Reviews, 58(1), 87–114.
  • Pashaei, H., Rouhani, A., Nejabat, M., Hadizadeh, F., Mirzaei, S., Nadri, H., Maleki, M. F., & Ghodsi, R. (2021). Synthesis and molecular dynamic simulation studies of novel N-(1-benzylpiperidin-4-yl) quinoline-4-carboxamides as potential acetylcholinesterase inhibitors. Journal of Molecular Structure, 1244, 130919. https://doi.org/10.1016/j.molstruc.2021.130919
  • Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., & Ferrin, T. E. (2004). UCSF Chimera—A visualization system for exploratory research and analysis. Journal of Computational Chemistry, 25(13), 1605–1612. https://doi.org/10.1002/jcc.20084
  • Rose, Y., Duarte, J. M., Lowe, R., Segura, J., Bi, C., Bhikadiya, C., Chen, L., Rose, A. S., Bittrich, S., Burley, S. K., & Westbrook, J. D. (2021). RCSB Protein Data Bank: Architectural advances towards integrated searching and efficient access to macromolecular structure data from the PDB archive. Journal of Molecular Biology, 433(11), 166704. https://doi.org/10.1016/j.jmb.2020.11.003
  • Sander, T., Freyss, J., von Korff, M., & Rufener, C. (2015). DataWarrior: An open-source program for chemistry aware data visualization and analysis. Journal of Chemical İnformation and Modeling, 55(2), 460–473. https://doi.org/10.1021/ci500588j
  • Sharma, J., Bhardwaj, V. K., Singh, R., Rajendran, V., Purohit, R., & Kumar, S. (2021). An in-silico evaluation of different bioactive molecules of tea for their inhibition potency against non structural protein-15 of SARS-CoV-2. Food Chemistry, 346, 128933. https://doi.org/10.1016/j.foodchem.2020.128933
  • Sharmaine, C., & Er, L. K. (2020). Xanthine oxidase ınhibitory activity of methanolic extract of Alternanthera sessilis. Sains Malaysiana, 49(2), 405–410.
  • Sheldrick, G. M. (2015a). SHELXT–Integrated space-group and crystal-structure determination. Acta Crystallographica. Section A, Foundations and Advances, 71(Pt 1), 3–8. https://doi.org/10.1107/S2053273314026370
  • Sheldrick, G. M. (2015b). Crystal structure refinement with SHELXL. Acta Crystallographica. Section C, Structural Chemistry, 71(Pt 1), 3–8. https://doi.org/10.1107/S2053229614024218
  • Sheppard, W. A. (1964). α-Fluorinated ethers. I. Aryl fluoroalkyl ethers1. The Journal of Organic Chemistry, 29(1), 1–11. https://doi.org/10.1021/jo01024a001
  • Sirajuddin, M., Ahmad, C., Khan, H., Ullahkhan, I., Tariq, M., & Ullah, N. (2022). Synthesis, spectroscopic characterization, biological screening and POM analysis of potentially bioactive copper (II) carboxylate complexes. Bulletin of the Chemical Society of Ethiopia, 36(1), 57–71. https://doi.org/10.4314/bcse.v36i1.6
  • Song, J. U., Jang, J. W., Kim, T. H., Park, H., Park, W. S., Jung, S. H., & Kim, G. T. (2016). Structure-based design and biological evaluation of novel 2-(indol-2-yl) thiazole derivatives as xanthine oxidase inhibitors. Bioorganic & Medicinal Chemistry Letters, 26(3), 950–954. https://doi.org/10.1016/j.bmcl.2015.12.055
  • Spek, A. L. (1990). PLATON, an integrated tool for the analysis of the results of a single crystal structure determination. Acta Crystallographica Section A: Foundations of Crystallography, 46(s1), c34–c34.
  • Talas, Z. S., Gok, Y., Ozdemir, I., Ates, B., Gunal, S., & Yilmaz, I. (2015). Synthesis, antioxidant and anti-microbial properties of two organoselenium compounds. Pakistan Journal of Pharmaceutical Sciences, 28(2), 611–616.
  • Talas, Z. S., Yilmaz, I., Ozdemir, I., Ates, B., Gok, Y., & Cetinkaya, B. (2009). Role of synthesized organoselenium compounds on protection of rat erythrocytes from DMBA-induced oxidative stress. Biological Trace Element Research, 128(2), 167–175.
  • Tezcan, B., Gök, Y., Sevinçek, R., Taslimi, P., Taskin‐Tok, T., Aktaş, A., Güzel, B., Aygün, M., & Gülçin, I. (2022). Benzimidazolium salts bearing the trifluoromethyl group as organofluorine compounds: Synthesis, characterization, crystal structure, in silico study, and inhibitory profiles against acetylcholinesterase and α‐glycosidase. Journal of Biochemical and Molecular Toxicology, 36(4), e23001. https://doi.org/10.1002/jbt.23001
  • Türkan, F., Atalar, M. N., Aras, A., Gülçin, İ., & Bursal, E. (2020). ICP-MS and HPLC analyses, enzyme inhibition and antioxidant potential of Achillea schischkinii Sosn. Bioorganic Chemistry, 94, 103333. https://doi.org/10.1016/j.bioorg.2019.103333
  • Uddin, N., Sirajuddin, M., Uddin, N., Tariq, M., Ullah, H., Ali, S., Tirmizi, S. A., & Khan, A. R. (2015). Synthesis, spectroscopic characterization, biological screenings, DNA binding study and POM analyses of transition metal carboxylates. Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy, 140, 563–574.
  • Wang, J., Sánchez-Roselló, M., Aceña, J. L., del Pozo, C., Sorochinsky, A. E., Fustero, S., Soloshonok, V. A., & Liu, H. (2014). Fluorine in pharmaceutical industry: Fluorine-containing drugs introduced to the market in the last decade (2001–2011). Chemical Reviews, 114(4), 2432–2506. https://doi.org/10.1021/cr4002879
  • Wanzlick, H. W., & Schönherr, H. J. (1968). Direct synthesis of a mercury salt‐carbene complex. Angewandte Chemie International Edition in English, 7(2), 141–142. https://doi.org/10.1002/anie.196801412
  • Whipker, B. E., McCall, I., Gibson, J. L., & Cavins, T. J. (2003). Efficacy of flurprimidol (Topflor) on bedding plants. Acta Horticulturae, 624(624), 413–418. https://doi.org/10.17660/ActaHortic.2003.624.58
  • Yadav, S., Deka, R., & Singh, H. B. (2019). Recent developments in the chemistry of NHC-based selones: Syntheses, applications and reactivity. Chemistry Letters, 48(2), 65–79. https://doi.org/10.1246/cl.180748
  • Yagiz, G., Noma, S. A. A., Altundas, A., Al-Khafaji, K., Taskin-Tok, T., & Ates, B. (2021). Synthesis, inhibition properties against xanthine oxidase and molecular docking studies of dimethyl N-benzyl-1H-1, 2, 3-triazole-4, 5-dicarboxylate and (N-benzyl-1H-1, 2, 3-triazole-4, 5-diyl) dimethanol derivatives. Bioorganic Chemistry, 108, 104654. https://doi.org/10.1016/j.bioorg.2021.104654
  • Yaqoob, M., Gul, S., Zubair, N. F., Iqbal, J., & Iqbal, M. A. (2020). Theoretical calculation of selenium N-heterocyclic carbene compounds through DFT studies: Synthesis, characterization and biological potential. Journal of Molecular Structure, 1204, 127462. https://doi.org/10.1016/j.molstruc.2019.127462
  • Yiğit, M., Celepci, D. B., Taslimi, P., Yiğit, B., Çetinkaya, E., Özdemir, İ., Aygün, M., & Gülçin, İ. (2022). Selenourea and thiourea derivatives of chiral and achiral enetetramines: Synthesis, characterization and enzyme inhibitory properties. Bioorganic Chemistry, 120, 105566. https://doi.org/10.1016/j.bioorg.2021.105566
  • Zhang, K., Zhao, P., Guo, G., Guo, Y., Tian, L., Sun, X., Li, S., He, Y., Sun, Y., Chai, H., Zhang, W., & Xing, M. (2016). Arsenic trioxide attenuates NF-κB and cytokine mRNA levels in the livers of cocks. Biological Trace Element Research, 170(2), 432–437. https://doi.org/10.1007/s12011-015-0455-8
  • Zhou, Y., Wang, J., Gu, Z., Wang, S., Zhu, W., Aceña, J. L., Soloshonok, V. A., Izawa, K., & Liu, H. (2016). Next generation of fluorine-containing pharmaceuticals, compounds currently in phase II–III clinical trials of major pharmaceutical companies: New structural trends and therapeutic areas. Chemical Reviews, 116(2), 422–518. https://doi.org/10.1021/acs.chemrev.5b00392

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.