5,380
Views
56
CrossRef citations to date
0
Altmetric
Editorial

Carbonic anhydrase inhibitors and their potential in a range of therapeutic areas

Pages 709-712 | Received 02 Sep 2018, Accepted 11 Sep 2018, Published online: 19 Sep 2018

The metalloenzyme carbonic anhydrase (CA, EC 4.2.1.1) is ubiquitously expressed in organisms all over the phylogenetic tree, with seven genetically distinct families known to date. By equilibrating CO2 and bicarbonate with generation of a H+ ion, CAs play a crucial role in pH regulation, in providing bicarbonate or H+ ions for electrolyte secretion, but also in several metabolic pathways such as lipogenesis, gluconeogenesis and ureagenesis. The CA inhibitors (CAIs) are clinically used as diuretics, anti-glaucoma agents and anti-epileptics, but novel applications in the management of cancer, neuropathic pain, sleep apnea, migraine, lowering intracranial pressure, and cerebral ischemia were recently reported. CA activators may be useful in the treatment of cognitive impairment and phobias. This special issue of Expert Opinion on Therapeutic Patents provides an update in these fields, with reviews dedicated to the applications in renal and central nervous system diseases, cancer and metastasis, as well as biomedical applications of prokaryotic CAs, widely abundant in pathogenic bacteria, but also in fungi and protozoa. Some patent evaluations for the use of CAIs in sleep apnea and regulation of mast cell response are also present in this special issue, demonstrating the many new fields in which modulators of the activity of these enzymes may have useful applications.

The CAs act as catalysts for the interconversion between CO2, bicarbonate, and protons, one of the simplest chemical reactions connected with vital processes [Citation1Citation6]. Indeed, CO2, a gas already present in the primeval earth atmosphere, is generated in most oxidative metabolic processes in all organisms (except chemolithoautotrophs), whereas plants use it for photosynthesis. This is probably the reason why at least seven genetically distinct CA families are known to date, being present in organisms all over the phylogenetic tree (denominated as α-, β-, γ-, δ-, ζ-, η-, and ɵ-CAs) [Citation7Citation15].

A large number of α-CA isoforms have been described in vertebrates: 15 in humans and other primates, and 16 in other mammals [Citation1Citation6]. Thus, α-CAs are so widespread enzymes probably because their role is connected to tightly controlled processes such as pH regulation and metabolism [Citation1Citation15]. The view of CAs as metabolic enzymes started to be considered recently, after their role in tumor metabolism has been understood in detail [Citation5,Citation6]. At least CA II, IX, and XII participate in pH regulation and metabolic processes within hypoxic tumors, in which the oxidative phosphorylation of glucose is impaired due to the low amount of oxygen present, and most energy is obtained by the alternative, glycolytic pathway, which leads to the formation of lactic acid [Citation5,Citation6,Citation16Citation19]. The mitochondrial isoform CA VA was also proven to be involved in biosynthetic processes such as lipogenesis, neoglucogenesis, ureagenesis among others [Citation20Citation22]. Thus, the clinical use of the CA inhibitors (CAIs) saw new developments in these areas in recent years. The sulfonamides incorporating primary SO2NH2 moieties [Citation23Citation33] were recognized to act as potent CAIs already in the 1950s, and the first diuretic based on this class of pharmacologic agents, acetazolamide (5-acetamido-1,3,4-thiadiazole-2-sulfonamide), started to be clinically used in 1956, with this drug still being used nowadays [Citation1]. In fact, the discovery of acetazolamide played an important role in the development of renal physiology and pharmacology and led to the discovery of many modern classes of diuretics [Citation30Citation33]. Aspects related to the renal applications of CAIs, alone or in combination with other pharmacological agents, are dealt with in the paper by Supuran in this special issue [Citation34]. Novel applications in the management of drug-induced renal injury were recently reported for classical CAIs such as acetazolamide, methazolamide and topiramate [Citation34], whereas the involvement of renal CA isoforms in the reabsorption of nitrite (NO2), which is the most abundant reservoir of the biologically highly potent signaling molecule nitric oxide (NO), has also been investigated in some details in the last years [Citation35,Citation36].

At least 9 of the various CA isoforms are present in the human central nervous system (CNS) and choroid plexus, but their functions are poorly understood, although it is clear that they are involved in crucial functions within these organs [Citation34]. CA inhibition in the CNS was exploited therapeutically for obtaining anticonvulsant agents already in the 1970s, with acetazolamide and methazolamide the most used such agents, in some forms of epilepsy [Citation37Citation39]. The mechanisms by which CAIs exert antiepileptic action are rather complex and there is not a definitive consensus among researchers on many aspects related to this pharmacological effect of drugs such as topiramate, zonisamide, or sulthiame [Citation37Citation40]. Inhibition of brain CAs leads to a diminished formation of bicarbonate and also changes the brain pH, contributing thus to an antiepileptic effect by several different pathways [Citation37Citation40]. Among the new applications recently claimed in the fields, were those of combination therapies of a CAI, preferentially topiramate, with other pharmacological agents, such as an aldosterone derivative, or benzodiazepines as a therapeutic option for obstructive sleep apnea, with several patents which are discussed in the Patent evaluation by Angeli and Supuran [Citation41]. There are several other pharmacologic applications of the CAIs related to the inhibition of CNS isoforms, among which idiopathic intracranial hypertension (IIH) [Citation42,Citation43], cerebral ischemia [Citation44], neuropathic pain [Citation45Citation47], and migraine [Citation48]. Drug design campaigns and the proof of concept study regarding the druggability of various CA isoforms for the management of these conditions were recently published [Citation43Citation47] and are discussed in detail in one of the reviews present in this special issue [Citation34].

CA activation was reported in an early stage of CA research, but the field remained highly controversial till recently, when work with highly purified enzymes and very precise techniques, such as the stopped-flow assay, undoubtedly demonstrated that the CA activators (CAAs) exist and that they take part to the catalytic cycle [Citation49Citation53]. However, up until recently there seem to be no pharmacological applications for the CAAs [Citation54]. Recently it has been unraveled that CAAs may be of great relevance in pathologies related to the pharmacological enhancement of synaptic efficacy, spatial learning, and memory, with the mechanistic details responsible for such phenomena reported by Blandina’s group [Citation54]. Administration of CAAs of the amino acid type was shown to lead to an enhanced spatial learning, which was antagonized by the simultaneous administration of a sulfonamide inhibitor, such as acetazolamide [Citation54]. Impairments of the fear memory consolidation were also observed in the course of such experiments, which might pave the way for pharmacologic applications in novel therapies for phobia and post-traumatic shock [Citation54]. The administration of the CAA rapidly activated the extracellular signal-regulated kinase (ERK) pathways, which is involved in a critical step of memory formation, both in the cortex and the hippocampus, the two brain areas involved in memory processing [Citation54]. Such interesting results might also lead to the use of CAAs for memory therapy in aging or in neurodegenerative conditions such as Alzheimer’s disease [Citation54].

A second review article from the Special issue deals with the inhibition of the tumor associated isoforms CA IX and XII, which may lead to novel therapies for the treatment of hypoxic, metastatic solid tumors [Citation55]. As mentioned above [Citation6,Citation16Citation19], several CA isoforms are predominantly present in tumors, in which they are involved in pH regulation and metabolic processes. It has been demonstrated that their specific inhibition with sulfonamides, sulfamates, or coumarins [Citation56Citation59] has profound effects on the inhibition of the primary tumor growth, metastases formation and reduction in the cancer stem cells number, three mechanisms unique only to this class of pharmacological agents [Citation60Citation62]. Thus, a large number of drug design studies were performed in the last 10 years for the design of CA IX/XII – selective inhibitors [Citation1,Citation3Citation6,Citation55], with one such compound, SLC-0111 presently in Phase Ib/II clinical trials as an antitumor/antimetastatic agent [Citation63]. All these aspects are dealt with in detail in the review by Nocentini and Supuran in this Special issue [Citation55].

A new aspect of CAI research is analyzed in the Patent evaluation by Winum [Citation64] published in the Special issue. It has recently been claimed that CA activity modulators (inhibitors or activators) may be also useful for the treatment of allergic diseases, viral, bacterial, or fungal infections, or mastocytosis, based on their effects on mast cell-mediated inflammation [Citation64].

The last review in the Special issue, by Supuran and Capasso [Citation65] deals with the biomedical applications of CAs of prokaryotic origin. As mentioned above [Citation1,Citation2,Citation4,Citation7], CAs from at least three classes (the α-, β- and γ-CA) are present in eukaryotes, in which they play crucial functions related to pH regulation, photosynthesis, acclimation to environmental conditions (e.g. extreme pH conditions, extreme temperatures, high concentrations of salts, etc. [Citation1,Citation2,Citation4,Citation7]), and metabolism [Citation2,Citation7]. Many representatives of such enzymes were cloned and characterized in detail from many pathogenic and nonpathogenic bacteria among which Helicobacter pylori, Vibrio cholerae, Brucella suis, Mycobacterium tuberculosis, etc. (to mention only the most relevant ones) [Citation66Citation72]. Successful drug design campaigns [Citation1,Citation2,Citation4,Citation7,Citation73] were also reported for many such enzymes from pathogenic bacteria in the search of agents able to overcome the extensive drug resistance observed with the classical antibiotics [Citation74]. However, there seems to be a lack of interest from drug companies for developing anti-infectives with an alternative mechanism of action based on CA inhibition. This review article brings the attention to these underexplored anti-infective drug targets, which, if properly considered, may represent a breakthrough to the field.

Overall, the very interesting and highly diverse topics treated in these review articles and Patent evaluations prove again, if necessary, the many aspects in which the CA research may bring highly innovative applications, in resolving challenging problems with which our evolving society is confronted nowadays.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This manuscript was not funded.

References

  • Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nature Rev Drug Discov. 2008;7:168–181.
  • Capasso C, Supuran CT. An overview of the alpha-, beta-and gamma-carbonic anhydrases from bacteria: can bacterial carbonic anhydrases shed new light on evolution of bacteria? J Enzyme Inhib Med Chem. 2015;30:325–332.
  • Alterio V, Di Fiore A, D’Ambrosio K, et al. Multiple binding modes of inhibitors to carbonic anhydrases: how to design specific drugs targeting 15 different isoforms? Chem Rev. 2012;112:4421–4468.
  • Supuran CT. Structure and function of carbonic anhydrases. Biochem J. 2016;473:2023–2032.
  • Supuran CT. Carbonic anhydrases and metabolism. Metabolites. 2018;8:E25.
  • Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov. 2011;10:767–777.
  • Capasso C, Supuran CT. Bacterial, fungal and protozoan carbonic anhydrases as drug targets. Expert Opin Ther Targets. 2015;19:1689–1704.
  • Supuran CT, Capasso C. New light on bacterial carbonic anhydrases phylogeny based on the analysis of signal peptide sequences. J Enzyme Inhib Med Chem. 2016;31:1254–1260.
  • Supuran CT, Capasso C. The eta-class carbonic anhydrases as drug targets for antimalarial agents. Expert Opin Ther Targets. 2015;19:551–563.
  • Supuran CT. Inhibition of carbonic anhydrase from Trypanosoma cruzi for the management of Chagas disease: an underexplored therapeutic opportunity. Future Med Chem. 2016;8:311–324.
  • Del Prete S, De Luca V, De Simone G, et al. Cloning, expression and purification of the complete domain of the η-carbonic anhydrase from Plasmodium falciparum. J Enzyme Inhib Med Chem. 2016;31(sup4):54–59.
  • Xu Y, Feng L, Jeffrey PD, et al. Structure and metal exchange in the cadmium carbonic anhydrase of marine diatoms. Nature. 2008;452:56–61.
  • Zimmerman SA, Ferry JG, Supuran CT. Inhibition of the archaeal β-class (Cab) and γ-class (Cam) carbonic anhydrases. Curr Top Med Chem. 2007;7:901–908.
  • Schlicker C, Hall RA, Vullo D, et al. Structure and inhibition of the CO2-sensing carbonic anhydrase Can2 from the pathogenic fungus Cryptococcus neoformans. J Mol Biol. 2009;385:1207–1220.
  • Del Prete S, De Luca V, Vullo D, et al. A new procedure for the cloning, expression and purification of the β-carbonic anhydrase from the pathogenic yeast Malassezia globosa, an anti-dandruff drug target. J Enzyme Inhib Med Chem. 2016;31:1156–1161.
  • Supuran CT. Carbonic anhydrase inhibition and the management of hypoxic tumors. Metabolites. 2017;7:E48.
  • Ward C, Langdon SP, Mullen P, et al. New strategies for targeting the hypoxic tumour microenvironment in breast cancer. Cancer Treat Rev. 2013;39:171–179.
  • Kusuzaki K, Matsubara T, Murata H, et al. Natural extracellular nanovesicles and photodynamic molecules: is there a future for drug delivery? J Enzyme Inhib Med Chem. 2017;32:908–916.
  • Iessi E, Logozzi M, Mizzoni D, et al. Rethinking the combination of proton exchanger inhibitors in cancer therapy. Metabolites. 2018;8:E2.
  • Arechederra RL, Waheed A, Sly WS, et al. Effect of sulfonamides as carbonic anhydrase VA and VB inhibitors on mitochondrial metabolic energy conversion. Bioorg Med Chem. 2013;21:1544–1548.
  • Salameh TS, Shah GN, Price TO, et al. Blood-brain barrier disruption and neurovascular unit dysfunction in diabetic mice: protection with the mitochondrial carbonic anhydrase inhibitor topiramate. J Pharmacol Exp Ther. 2016;359:452–459.
  • Price TO, Sheibani N, Shah GN. Regulation of high glucose-induced apoptosis of brain pericytes by mitochondrial CA VA: A specific target for prevention of diabetic cerebrovascular pathology. Biochim Biophys Acta. 2017;1863:929–935.
  • Supuran CT. How many carbonic anhydrase inhibition mechanisms exist? J Enzyme Inhib Med Chem. 2016;31:345–360.
  • Supuran CT, Scozzafava A. Carbonic anhydrase inhibitors and their therapeutic potential. Expert Opin Ther Pat. 2000;10:575–600.
  • Supuran CT. Structure-based drug discovery of carbonic anhydrase inhibitors. J Enzyme Inhib Med Chem. 2012;27:759–772.
  • Supuran CT. Advances in structure-based drug discovery of carbonic anhydrase inhibitors. Expert Opin Drug Discov. 2017;12:61–88.
  • Carta F, Supuran CT. Diuretics with carbonic anhydrase inhibitory action: a patent and literature review (2005-2013). Expert Opin. Ther. Pat. 2013;23:681–691.
  • Masini E, Carta F, Scozzafava A, et al. Antiglaucoma carbonic anhydrase inhibitors: a patent review. Expert Opin. Ther. Pat. 2013;23:705–716.
  • Scozzafava A, Supuran CT, Carta F. Antiobesity carbonic anhydrase inhibitors: a literature and patent review. Expert Opin. Ther. Pat. 2013;23:725–735.
  • Monti SM, Supuran CT, De Simone G. Anticancer carbonic anhydrase inhibitors: a patent review (2008-2013). Expert Opin Ther Pat. 2013;23:737–749.
  • Carta F, Scozzafava A, Supuran CT. Sulfonamides (RSO2NH2): a patent review 2008-2012. Expert Opin Ther Pat. 2012;22:747–758.
  • Scozzafava A, Carta F, Supuran CT. Secondary and tertiary sulfonamides: a patent review (2008-2012). Expert Opin Ther Pat. 2013;23:203–213.
  • Carta F, Supuran CT, Scozzafava A. Novel therapies for glaucoma: a patent review 2007-2011. Expert Opin. Ther. Pat. 2012;22:79–88.
  • Supuran CT. Applications of carbonic anhydrases inhibitors in renal and central nervous system diseases. Expert Opin Ther Pat. Forthcoming 2018:1–9. DOI:10.1080/13543776.2018.1519023
  • Hanff E, Zinke M, Böhmer A, et al. GC-MS determination of nitrous anhydrase activity of bovine and human carbonic anhydrase II and IV. Anal Biochem. 2018;550:132–136.
  • Chobanyan-Jürgens K, Schwarz A, Böhmer A, et al. Renal carbonic anhydrases are involved in the reabsorption of endogenous nitrite. Nitric Oxide. 2012;26:126–131.
  • Thiry A, Dognè JM, Masereel B, et al. Carbonic anhydrase inhibitors as anticonvulsant agents. Curr Top Med Chem. 2007;7:855–864.
  • Aggarwal M, Kondeti B, McKenna R. Anticonvulsant/antiepileptic carbonic anhydrase inhibitors: a patent review. Expert Opin Ther Pat. 2013;23:717–724.
  • Hamidi S, Avoli M. Carbonic anhydrase inhibition by acetazolamide reduces in vitro epileptiform synchronization. Neuropharmacology. 2015;95:377–387.
  • Casini A, Antel J, Abbate F, et al. Carbonic anhydrase inhibitors: SAR and X-ray crystallographic study for the interaction of sugar sulfamates/sulfamides with isozymes I, II and IV. Bioorg Med Chem Lett. 2003;13:841–845.
  • Angeli A, Supuran CT. Treatment of sleep apnea with a combination of a carbonic anhydrase inhibitor and an aldosterone antagonist: a patent evaluation of CA2958110 and IN6616DEN2012. Expert Opin Ther Pat. Forthcoming 2018:1–5. DOI:10.1080/13543776.2018.1497160
  • Supuran CT. Acetazolamide for the treatment of idiopathic intracranial hypertension. Expert Rev Neurother. 2015;15:851–856.
  • Supuran CT. Drug interaction considerations in the therapeutic use of carbonic anhydrase inhibitors. Expert Opin Drug Metab Toxicol. 2016;12:423–431.
  • Di Cesare Mannelli L, Micheli L, Carta F, et al. Carbonic anhydrase inhibition for the management of cerebral ischemia: in vivo evaluation of sulfonamide and coumarin inhibitors. Enzyme Inhib Med Chem. 2016;31:894–899.
  • Asides M, Ossipov MH, Kaila K, et al. Acetazolamide and midazolam act synergistically to inhibit neuropathic pain. Pain. 2010;148:302–308.
  • Carta F, Di Cesare Mannelli L, Pinard M, et al. A class of sulfonamide carbonic anhydrase inhibitors with neuropathic pain modulating effects. Bioorg Med Chem. 2015;23:1828–1840.
  • Supuran CT. Carbonic anhydrase inhibition and the management of neuropathic pain. Expert Rev Neurother. 2016;16:961–968.
  • Silberstein SD. Topiramate in migraine prevention: a 2016 perspective. Headache. 2017;57:165–178.
  • Supuran CT. Carbonic anhydrase activators. Future Med Chem. 2018;10:561–573.
  • Briganti F, Mangani S, Orioli P, et al. Carbonic anhydrase activators: X-ray crystallographic and spectroscopic investigations for the interaction of isozymes I and II with histamine. Biochemistry. 1997;36:10384–10392.
  • Temperini C, Scozzafava A, Vullo D, et al. Carbonic anhydrase activators. Activation of isozymes I, II, IV, VA, VII, and XIV with l- and d-histidine and crystallographic analysis of their adducts with isoform II: engineering proton-transfer processes within the active site of an enzyme. Chemistry. 2006;12:7057–7066.
  • Temperini C, Scozzafava A, Vullo D, et al. Carbonic anhydrase activators. Activation of isoforms I, II, IV, VA, VII, and XIV with L- and D-phenylalanine and crystallographic analysis of their adducts with isozyme II: stereospecific recognition within the active site of an enzyme and its consequences for the drug design. J Med Chem. 2006;49:3019–3027.
  • Angeli A, Vaiano F, Mari F, et al. Psychoactive substances belonging to the amphetamine class potently activate brain carbonic anhydrase isoforms VA, VB, VII, and XII. J Enzyme Inhib Med Chem. 2017;32:1253–1259.
  • Canto de Souza L, Provensi G, Vullo D, et al. Carbonic anhydrase activation enhances object recognition memory in mice through phosphorylation of the extracellular signal-regulated kinase in the cortex and the hippocampus. Neuropharmacology. 2017;118:148–156.
  • Nocentini A, Supuran CT. Carbonic anhydrase inhibitors as antitumor/antimetastatic agents: a patent review (2008-2018). Expert Opin Ther Pat. 2018;1–12. DOI:10.1080/13543776.2018.1508453
  • Pacchiano F, Carta F, McDonald PC, et al. Ureido-substituted benzenesulfonamides potently inhibit carbonic anhydrase IX and show antimetastatic activity in amodel of breast cancer metastasis. J Med Chem. 2011;54:1896–1902.
  • Gieling RG, Babur M, Mamnani L, et al. Antimetastatic effect of sulfamate carbonic anhydrase IX inhibitors in breast carcinoma xenografts. J Med Chem. 2012;55:5591–5600.
  • Krall N, Pretto F, Decurtins W, et al. A small-molecule drug conjugate for the treatment of carbonic anhydrase IX expressing tumors. Angew Chem Int Ed Engl. 2014;53:4231–4235.
  • Ferraroni M, Carta F, Scozzafava A, et al. Thioxocoumarins show an alternative carbonic anhydrase inhibition mechanism compared to coumarins. J Med Chem. 2016;59:462–473.
  • Lou Y, McDonald PC, Oloumi A, et al. Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Res. 2011;71:3364–3376.
  • Bozdag M, Carta F, Ceruso M, et al. Discovery of 4-hydroxy-3-(3-(phenylureido)benzenesulfonamides as SLC-0111 analogues for the treatment of hypoxic tumors overexpressing carbonic anhydrase IX. J Med Chem. 2018;61:6328–6338.
  • Lock FE, McDonald PC, Lou Y, et al. Targeting carbonic anhydrase IX depletes breast cancer stem cells within the hypoxic niche. Oncogene. 2013;32:5210–5219.
  • [cited 2018 Sep 2]. Available from: https://clinicaltrials.gov/ct2/show/NCT03450018.
  • Winum JY. Carbonic anhydrase enzymes for regulating mast cell hematopoiesis and type-2 inflammation: a patent evaluation (WO2017/058370). Expert Opin Ther Pat. Forthcoming 2018:1–3. DOI:10.1080/13543776.2018.1501472
  • Supuran CT, Capasso C. Biomedical applications of prokaryotic carbonic anhydrases. Expert Opin Ther Pat. 2018;1–10. DOI:10.1080/13543776.2018.1497161
  • Nishimori I, Minakuchi T, Morimoto K, et al. Carbonic anhydrase inhibitors: DNA cloning and inhibition studies of the alpha-carbonic anhydrase from Helicobacter pylori, a new target for developing sulfonamide and sulfamate gastric drugs. J Med Chem. 2006;49:2117–2126.
  • Nishimori I, Onishi S, Takeuchi H, et al. The alpha and beta classes carbonic anhydrases from helicobacter pylori as novel drug targets. Curr Pharm Des. 2008;14:622–630.
  • Joseph P, Ouahrani-Bettache S, Montero JL, et al. A new beta-carbonic anhydrase from Brucella suis, its cloning, characterization, and inhibition with sulfonamides and sulfamates, leading to impaired pathogen growth. Bioorg Med Chem. 2011;19:1172–1178.
  • Del Prete S, Isik S, Vullo D, et al. DNA cloning, characterization, and inhibition studies of an alpha-carbonic anhydrase from the pathogenic bacterium Vibrio cholerae. J Med Chem. 2012;55:10742–10748.
  • Güzel O, Maresca A, Scozzafava A, et al. Discovery of low nanomolar and subnanomolar inhibitors of the mycobacterial beta-carbonic anhydrases Rv1284 and Rv3273. J Med Chem. 2009;52:4063–4067.
  • Nishimori I, Minakuchi T, Vullo D, et al. Carbonic anhydrase inhibitors. Cloning, characterization, and inhibition studies of a new beta-carbonic anhydrase from Mycobacterium tuberculosis. J Med Chem. 2009;52:3116–3120.
  • Nishimori I, Minakuchi T, Maresca A, et al. The β-carbonic anhydrases from Mycobacterium tuberculosis as drug targets. Curr Pharm Des. 2010;16:3300–3309.
  • Supuran CT. Carbon- versus sulphur-based zinc binding groups for carbonic anhydrase inhibitors? J Enzyme Inhib Med Chem. 2018;33:485–495.
  • Supuran CT. Inhibition of bacterial carbonic anhydrases and zinc proteases: from orphan targets to innovative new antibiotic drugs. Curr Med Chem. 2012;19:831–844.

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.