2,714
Views
14
CrossRef citations to date
0
Altmetric
Research Papers

Antiproliferative effects of sulphonamide carbonic anhydrase inhibitors C18, SLC-0111 and acetazolamide on bladder, glioblastoma and pancreatic cancer cell lines

, , , ORCID Icon, ORCID Icon &
Pages 280-286 | Received 10 Oct 2021, Accepted 05 Nov 2021, Published online: 11 Dec 2021

Abstract

Carbonic anhydrase IX/XII (CA IX/XII), are cell-surface enzymes typically expressed by cancer cells as a form of adaptation to hypoxia and acidosis. It has been widely reported that these proteins play pivotal roles in cancer progression fostering cell migration, aggressiveness and resistance to first line chemo- and radiotherapies. CA IX has emerged as a promising target in cancer therapy and several approaches and families of compounds were characterised in the attempt to find optimal targeting by inhibiting of the high catalytic activity of the enzyme. In the present work, different cell lines representing glioblastoma, bladder and pancreatic cancer have been exploited to compare the inhibitory and antiproliferative effect of primary sulphonamide acetazolamide (AAZ), the Phase Ib/II clinical grade sulphonamide SLC-0111, and a membrane-impermeant positively charged, pyridinium-derivative (C18). New hints regarding the possibility to exploit CA inhibitors in these cancer types are proposed.

1. Introduction

In the tumour contexts, the rapid and uncontrolled proliferation of neoplastic cells is mainly limited by the availability of oxygen, due to the insufficiency of blood supply. Hypoxia thus represents a typical feature of the majority of solid tumours. It has been reported that chronic (but also cycling) hypoxia leads to a range of diverse phenomena, among which increased formation of reactive oxygen species (ROS) contributing to increased genetic instability and mutagenesisCitation1,Citation2, that leads to tumour cell survival and progressionCitation3. In addition, the hypoxic milieu has been associated with increased resistance to the metastatic phenotype and resistance to radiation therapy and chemotherapyCitation4–8. Due to the high proliferative rate and metabolic turnover, cancer cells exploit the oxidative phosphorylation of glucose and its anaerobic glycolysis for energy production, thus generating high quantities of metabolic acids that require the intervention of a various protein complexes involved in the prevention of intracellular accumulation of H+ ions, and the maintenance of an intracellular alkaline pHCitation3,Citation4. In this context, the hypoxia-inducible factor-1 (HIF-1) represents the main transcription factor activated in response to hypoxia and driving the regulations of many cellular biological processes including glycolysis, cell migration, angiogenesis, and pH regulationCitation1–4. In particular, HIF-1 can regulate the expression and activation of various pH-regulating proteins, such as sodium bicarbonate transporters, sodium-proton exchangers, monocarboxylate transporter-4 (MCT-4), and the carbonic anhydrase IX/XII (CA IX/XII) isoforms, that lead to an inverted pH gradient, typical of cancer cellsCitation9, i.e. acidic extracellular pH and slightly more alkaline cytosolic pH values compared to normal ones.

CA IX and XII are cell-surface enzymes overexpressed in hypoxia, belonging to the α-CA (EC 4.2.1.1) family of zinc metalloenzymes, and catalyse the reversible hydration of CO2 to bicarbonate ions (HCO3) and protons (H+)Citation10. These enzymes are involved in the adaptation of cancer cells to acidosis, but are also involved in ferroptosis and several other processes by which tumour cells escape control and are able to proliferateCitation10. High levels of CA IX/XII have been reported in several types of cancer where it promotes cell survival under hypoxic conditions and may favour tumour cells migration and aggressivenessCitation11–13. Moreover, the expression of CA IX has been correlated with poor prognosis in breast, ovarian, pancreatic, bladder, brain and other human tumoursCitation14,Citation15, and its targeting has been widely proposed as a therapeutic approach to treat aggressive cancersCitation16. To this purpose different inhibitors have been developed and characterised acting as blockers of the CA IX activityCitation3,Citation4, including the sulphonamides, sulphonamide derivatives, other small molecules inhibitors, and antibody/cytokine-drug conjugatesCitation17,Citation18. In the present work we compare the in vitro effect of different CA IX inhibitors on various tumour types. In particular, we compare the biological activity of the primary sulphonamide acetazolamide (AAZ)Citation18, the sulphonamide SLC-0111, actually in Phase Ib/II clinical trials for the management of advanced solid tumoursCitation19, and a positively charged, pyridinium-derivative (C18) characterised by the fact that it is membrane impermeantCitation20.

2. Materials and methods

2.1. Cell culture and reagents

Human bladder cancer RT4 cells were obtained from ATCC-LGC Standards Repository (ATCC number HTB-2) and maintained in McCoy’s 5 A medium supplemented with 10% heat-inactivated FCS. Human bladder cancer 5637 cells (ATCC HTB-9), Human pancreatic cancer CF-PAC-1 (ATCC CRL-1918) and PANC-1 (ATCC CRL-1469) cells were grown in RPMI 1640 supplemented with 10% FCS. Human bladder cancer HT-137 cells (ATCC CRL-1472), human glioblastoma U87MG (ATCC HTB-14), U251 (Merck U-251 MG), T98G (ATCC CRL-1690) cells were grown in DMEM supplemented with 10% FCS. Cells were kept at low passage, returning to original frozen stocks every 3 to 4 months. Hypoxic culture conditions were realised in the presence of 1% O2 e 5% CO2. AAZ (acetazolamide) was commercially available from Sigma-Aldrich (Milan, Italy), whereas SLC-0111Citation21 and C18Citation20 were prepared as reported earlierCitation20,Citation21.

2.2. Western blot analysis

Cells were cultured at 37 °C with 1% O2, 5% CO2 for 24 h and lysed in lysis buffer (TRIS-HCl pH 7 50 mM, NaCl 150 mM, Triton X-100 1%, BriJ 0.1%). Protein concentrations were determined using the Bradford protein assay (Bio-Rad Laboratories, Milano, Italy). Then, 60 μg protein/sample were separated by SDS-PAGE, analysed by WB for CA9 with the murine mAb M75 an IgG2b and normalised with an anti-GAPDH (Santa Cruz Biotechnology).

2.3. Cell proliferation assay

The different cellular lines were seeded in 48-well plates and treated in 1% FBS with increasing concentrations of AAZ, SLC-001 or C18. After 72 h of incubation at 37 °C with 1% O2, 5% CO2, cells were trypsinized and cell counting was performed with the MACSQuant® Analyzer (Miltenyi Biotec).

3. Results and discussion

3.1. Ca inhibitory effects of sulphonamides C18, SLC-0111 and acetazolamide

Sulphonamides constitute one of the most investigated class of CA inhibitors (CAIs), with many such compounds on clinical use for decades for the management of many diseases connected with CA imbalances, or more recently, in clinical trials as antitumor agentsCitation16,Citation17.

We decided to investigate here three such CAIs with very different properties and CA inhibition profiles against the two human (h) hCA isoforms involved in tumorigenesis ( and ), hCA IX and XII, as well as the two main off-target isoforms, the cytosolic hCA I and IICitation3,Citation16,Citation17. Indeed, compound 1, also known as C18, belongs to a class of positively-charged, membrane-impermeant compoundsCitation23 that were demonstrated to be unable to cross plasma membranes and thus selectively inhibit transmembrane isoforms such as CA IX and XIICitation23,Citation24. C18 is an effective, low nanomolar inhibitor of CA II, IX and XII, being a weak inhibitor against hCA I (see andCitation16) However, due to its membrane-impermeability, in vivo it should predominantly inhibit the transmembrane isoforms, such as CA IX and XIICitation23. SLC-0111, compound 2, as mentioned above, has been designed by the tail approach as a selective CAI for the tumour-associated isoformsCitation21. The compound is a low nanomolar CA IX/XII inhibitor, whereas its activity against the off-target isoforms hCA I and II is in the micromolar range ( andCitation21) Acetazolamide 3, is the CAI par excellence, being in clinical use for almost 70 years as a diuretic, antiglaucoma and antiepileptic agentCitation16, being also employed for the management of other conditions such as mountain sickness, idiopathic intracranial hypertension, and other CNS or renal conditionsCitation16,Citation17,Citation25. As seen from data of , AAZ is a promiscuous, highly effective CAI against all the 4 isoforms considered here, CA I, II, IX and XII.

Figure 1. Sulphonamides 1–3 investigated in this work, C18, SLC-0111 and AAZ.

Figure 1. Sulphonamides 1–3 investigated in this work, C18, SLC-0111 and AAZ.

Table 1. CA inhibition data with sulphonamides 1–3 against human CA isoforms of clinical relevance.

3.2. Evaluation of sulphonamide CAIs effects on bladder cancer cells

We evaluated the effect of AAZ, SLC-0111 and C18 on three tumour cell lines representing different grades of bladder cancer progression: low grade/papilloma-like RT4 cells, grade II 5637 cells, and grade III/muscle invasive HT-1376 cells. Firstly, we analysed CAIX expression under hypoxic conditions. As shown in , all the bladder cancer cells express basal levels of CAIX that significantly rise after exposure to hypoxic conditions for 24 h. We compared the efficacy of the three CAIX inhibitors to inhibit the proliferation of these after 72 h of treatment in hypoxia. As shown in , both the AAZ had no effect on cell proliferation on all cell lines. Similarly, the membrane impermeant C18 compound had a very mild effect on proliferation of medium (5637) and high-grade cells (HT-1376), but only at high concentrations. Finally, the clinical grade SLC-0111 drug displayed a modest inhibitory effect on RT4 and 5637 cells, but not on HT-1376 cells.

Figure 2. Protein levels of CA IX in bladder cancer cells under normal (NT) or hypoxic (HYPO) conditions (top panels), and cell proliferation of these cells treated with C18, SLC-0111 or AAZ.

Figure 2. Protein levels of CA IX in bladder cancer cells under normal (NT) or hypoxic (HYPO) conditions (top panels), and cell proliferation of these cells treated with C18, SLC-0111 or AAZ.

These data suggest that bladder cancer cells, regardless from their grade and aggressiveness are not particularly sensitive to the inhibition of CA IX.

3.3. Evaluation of sulphonamide CAI effects on glioblastoma cells

The expression of CAIX was evaluated by Western blot in three glioblastoma cell lines maintained for 24 h in hypoxic conditions. As shown in , CA IX levels are significantly increased after exposure to hypoxia for 24 h. We then evaluated the anti-proliferative effect of the three CA IX inhibitors under these conditions. No effect was observed when cells were treated with AAZ for 72 h, while a mild effect was observed after treatment with C18 in U251 and in T98G cells. Notably, the clinical grade SLC-0111 compound showed a significant effect on all the three cell lines, with an IC50 ranging from 80 to 100 µM. These data suggest that CA IX blockade can significantly impact on glioblastoma cells, and that the loss of impermeant C18 drug has a mild efficacy in comparison with SLC-0111, but a better profile in respect to AAZ.

Figure 3. Protein levels of CA IX in glioblastoma cells under normal (NT) or hypoxic (HYPO) conditions (top panels), and cell proliferation of these cells treated with C18, SLC-0111 or AAZ.

Figure 3. Protein levels of CA IX in glioblastoma cells under normal (NT) or hypoxic (HYPO) conditions (top panels), and cell proliferation of these cells treated with C18, SLC-0111 or AAZ.

3.4. Evaluation of sulphonamides effects on pancreatic cancer cells

In line with our previous experiments, we confirmed the expression of CA IX in two cell lines of pancreatic cancer (CF-PAC-1 and PANC-1 cells) under hypoxic conditions (. As shown in we treatment with AAZ slightly inhibited the proliferation only in CF-PAC-1 cells with no effect on the proliferation of PANC-1 cells even at very high concentrations. Interestingly, the membrane impermeant C18 compound was able to reduce cell proliferation in CF-PAC-1 cells, with an IC50≈85 µM and a very modest effect on PANC-1 cells. Finally, SLC-0111 impaired cell proliferation in both pancreatic cell lines with an IC50 ≈120–125 µM. These data point to a promising effect of CA IX inhibition in pancreatic cancer cells and show a slight effect of C18 compound in respect to what observed in other cancer models.

Figure 4. Protein levels of CA IX in pancreatic cancer cells under normal (NT) or hypoxic (HYPO) conditions (top panels), and cell proliferation of these cells treated with C18, SLC-0111 or AAZ.

Figure 4. Protein levels of CA IX in pancreatic cancer cells under normal (NT) or hypoxic (HYPO) conditions (top panels), and cell proliferation of these cells treated with C18, SLC-0111 or AAZ.

4. Conclusions

Sulphonamide CAIs started to be investigated as potential anti-tumour agents in the last two decades after the discovery of the two tumour-associated isoforms CA IX and XII, and their upregulation in hypoxiaCitation3,Citation4. Nowadays, a large number of such derivatives were characterised in detail regarding their in vitro inhibitory profile against the target and off-target isoforms, but also considering pharmacokinetic and pharmacodynamics propertiesCitation26. In fact, although many low nanomolar in vitro CAIs were reported, in cellular systems or in vivo, in animal models, not all these compounds showed the expected anti-proliferative activityCitation26. Although such effects are reported for most classes of antitumor agents, here we evaluated three sulphonamide CAIs with very different inhibition profiles and physico-chemical properties in order to possibly address this conundrum: a positively-charged, membrane-impermeant compound (C18) with a rather selective CA IX/XII inhibition; the drug candidate in Phase Ib/II clinical trials SLC-0111 which is a CA IX/XII-selective inhibitor, and the pan-inhibitor acetazolamide. All these three compounds are highly effective against the target enzymes CA IX and XII, but they possess variable activity against the off-target isoforms (). The data obtained against several tumour cell lines expressing variable amounts of CA IX reported here, indeed show a quite different antiproliferative activity of the three compounds. For instance, bladder cancer cells representing different grade of this urological neoplasia are mostly insensitive to all CAIs tested, apart from SLC-0111 that showed only a modest effect in the medium grade invasive 5637 cells. Interestingly, in the more aggressive and invasive cancer types, represented by glioblastoma and pancreatic cancer cells, the clinical candidate SLC-0111 exerted a significant antiproliferative effect. These results were confirmed in all the five cell lines tested and suggest that in these highly proliferating cancers the inhibition of CA IX might provide a beneficial therapeutic effect. In addition, when the membrane impermeant compound C18 was tested in these tumour cells, its activity was generally poor in glioblastoma cells, but a promising activity was observed in one of the pancreatic cancer lines.

In general, our data suggest that more aggressive tumours might be more sensitive to CA IX inhibition and this should be taken into consideration in addition to the simple presence/expression of the target. Indeed, even though in vivo validation of these observations will be required, highly proliferative tumours are expected to be more prone to generate a hypoxic environment and thus, be more sensitive to CA blockers. As shown here, the effect and role of CA IX has been reported in different tumour settings and, even if the in vivo data clearly reveal the key role of these proteins in tumour progression, resistance to therapy and promotion of tumour invasion, extensive in vitro data showed that CA IX inhibition may affect tumour cell survival and proliferationCitation27 and/or motility and invasionCitation28, dependently on the tumour cell type. In this work we focussed only on the proliferative outcome of CA IX inhibition in order to evaluate the effect of the membrane-impermeant compound C18 in comparison with the more permeable derivatives SLC-0111 and AAZ, showing a rather different anti-proliferative outcome. Finally, as already reported for other tumour typesCitation3,Citation4,Citation27–31, it will be extremely interesting to validate the efficacy of these compounds in combination with conventional and/or new chemotherapeutic treatments in order to assess if CA inhibition may improve the response or prevent the onset of resistance to these therapies.

Disclosure statement

CT Supuran is Editor-in-Chief of Journal of Enzyme Inhibition and Medicinal Chemistry and he was not involved in the assessment, peer review or decision-making process of this paper. The authors have no relevant affiliations of financial involvement with any organisation 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.

Additional information

Funding

This work was financed in part by an Italian Ministry for Research (MIUR) grant to CTS [PRIN: rot. 2017XYBP2R], and by Associazione Italiana per la Ricerca sul Cancro [AIRC IG 2019 – ID. 23151 to R.R.].

References

  • a) Chan N, Koritzinsky M, Zhao H, et al. Chronic hypoxia decreases synthesis of homologous recombination proteins to offset chemoresistance and radioresistance. Cancer Res 2008;68:605–14. b) Kondo A, Safaei R, Mishima M, et al. Hypoxia-induced enrichment and mutagenesis of cells that have lost DNA mismatch repair. Cancer Res 2001;61:7603–7.c) Semenza GL. Intratumoral hypoxia and mechanisms of immune evasion mediated by hypoxia-inducible factors. Physiology 2021;36:73–83.
  • a) Hsieh CH, Lee CH, Liang JA, et al. Cycling hypoxia increases U87 glioma cell radioresistance via ROS induced higher and long-term HIF-1 signal transduction activity. Oncol Rep 2010;24:1629–36. b) Kato Y, Yashiro M, Fuyuhiro Y, et al. Effects of acute and chronic hypoxia on the radiosensitivity of gastric and esophageal cancer cells. Anticancer Res 2011;31:3369–75. c) Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999; 399:271–5.
  • a) McDonald PC, Swayampakula M, Dedhar S. Coordinated regulation of metabolic transporters and migration/invasion by carbonic anhydrase IX. Metabolites 2018;8:20. b) Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov 2011;10:767–77. c) Supuran CT. Carbonic anhydrase inhibitors as emerging agents for the treatment and imaging of hypoxic tumors. Expert Opin Investig Drugs 2018;27:963–70. d) 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;71:3364–76.
  • a) Angeli A, Carta F, Nocentini A, et al. Carbonic anhydrase inhibitors targeting metabolism and tumor microenvironment. Metabolites 2020;10:412. b) Supuran CT. Carbonic anhydrase inhibition and the management of hypoxic tumors. Metabolites 2017;7:48. c) Supuran CT, Alterio V, Di Fiore A, et al. Inhibition of carbonic anhydrase IX targets primary tumors, metastases, and cancer stem cells: three for the price of one. Med Res Rev 2018;38:1799–836.
  • Chaplin DJ, Durand RE, Olive PL. Acute hypoxia in tumors: implications for modifiers of radiation effects. Int J Radiat Oncol Biol Phys 1986;12:1279–82.
  • Rofstad EK, Gaustad JV, Egeland TA, et al. Tumors exposed to acute cyclic hypoxic stress show enhanced angiogenesis, perfusion and metastatic dissemination. Int J Cancer 2010;127:1535–46.
  • Minassian LM, Cotechini T, Huitema E, Graham CH. Hypoxia-induced resistance to chemotherapy in cancer. Adv Exp Med Biol 2019;1136:123–39.
  • Samanta D, Gilkes DM, Chaturvedi P, et al. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc Natl Acad Sci U S A 2014;111:E5429–38.
  • Swietach P. What is pH regulation, and why do cancer cells need it? Cancer Metastasis Rev 2019;38:5–15.
  • a) Supuran CT. Structure and function of carbonic anhydrases. Biochem J 2016;473: 2023–32. b) Chafe SC, Vizeacoumar FS, Venkateswaran G, et al. Genome-wide synthetic lethal screen unveils novel CAIX-NFS1/xCT axis as a targetable vulnerability in hypoxic solid tumors. Sci Adv. 2021;7:eabj0364.
  • Güttler A, Theuerkorn K, Riemann A, et al. Cellular and radiobiological effects of carbonic anhydrase IX in human breast cancer cells. Oncol Rep 2019;41:2585–94.
  • a) Koyuncu I, Tülüce Y, Slahaddin Qadir H, et al. Evaluation of the anticancer potential of a sulphonamide carbonic anhydrase IX inhibitor on cervical cancer cells. J Enzyme Inhib Med Chem 2019;34:703–11. b) Koyuncu I, Gonel A, Durgun M, et al. Assessment of the antiproliferative and apoptotic roles of sulfonamide carbonic anhydrase IX inhibitors in HeLa cancer cell line. J Enzyme Inhib Med Chem 2019;34:75–86.
  • Hsin MC, Hsieh YH, Hsiao YH, et al. Carbonic anhydrase IX promotes human cervical cancer cell motility by regulating PFKFB4 expression. Cancers 2021;13:1174.
  • Chen CL, Chu JS, Su WC, et al. Hypoxia and metabolic phenotypes during breast carcinogenesis: expression of HIF-1alpha, GLUT1, and CAIX. Virchows Arch 2010;457:53–61.
  • a) Andring JT, Fouch M, Akocak S, et al. Structural basis of nanomolar inhibition of tumor-associated carbonic anhydrase IX: X-ray crystallographic and inhibition study of lipophilic inhibitors with acetazolamide backbone. J Med Chem 2020;63:13064–75. b) Briganti F, Pierattelli R, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Part 37. Novel classes of carbonic anhydrase inhibitors and their interaction with the native and cobalt-substituted enzyme: kinetic and spectroscopic investigations. Eur J Med Chem 1996;31:1001–10. c) Carta F, Aggarwal M, Maresca A, et al. Dithiocarbamates: a new class of carbonic anhydrase inhibitors. Crystallographic and kinetic investigations. Chem Commun 2012;48:1868–70.
  • Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov 2008;7:168–81.
  • Supuran CT. Experimental carbonic anhydrase inhibitors for the treatment of hypoxic tumors. J Exp Pharmacol 2020;12:603–17.
  • Supuran CT. Emerging role of carbonic anhydrase inhibitors. Clin Sci 2021;135:1233–49.
  • McDonald PC, Chia S, Bedard PL, et al. A phase 1 study of SLC-0111, a novel inhibitor of carbonic anhydrase IX, in patients with advanced solid tumors. Am J Clin Oncol 2020;43:484–90.
  • a) Nocentini A, Angeli A, Carta F, et al. Reconsidering anion inhibitors in the general context of drug design studies of modulators of activity of the classical enzyme carbonic anhydrase. J Enzyme Inhib Med Chem 2021;36:561–80. b) Menchise V, De Simone G, Alterio V, et al. Carbonic anhydrase inhibitors: stacking with Phe131 determines active site binding region of inhibitors as exemplified by the X-ray crystal structure of a membrane-impermeant antitumor sulfonamide complexed with isozyme II. J Med Chem 2005;48:5721–7.
  • a) Pacchiano F, Carta F, McDonald PC, et al. Ureido-substituted benzenesulfonamides potently inhibit carbonic anhydrase IX and show antimetastatic activity in a model of breast cancer metastasis. J Med Chem 2011;54:1896–902. b) Pacchiano F, Aggarwal M, Avvaru BS, et al. Selective hydrophobic pocket binding observed within the carbonic anhydrase II active site accommodate different 4-substituted-ureido-benzenesulfonamides and correlate to inhibitor potency. Chem Commun 2010;46:8371–3.
  • Khalifah RG. The carbon dioxide hydration activity of carbonic anhydrase. I. Stop-flow kinetic studies on the native human isoenzymes B and c. J Biol Chem 1971;246:2561–257.
  • Supuran CT, Scozzafava A. Benzolamide is not a membrane-impermeant carbonic anhydrase inhibitor. J Enzyme Inhib Med Chem 2004;19:269–73.
  • a) Scozzafava A, Briganti F, Ilies MA, Supuran CT. Carbonic anhydrase inhibitors: synthesis of membrane-impermeant low molecular weight sulfonamides possessing in vivo selectivity for the membrane-bound versus cytosolic isozymes. J Med Chem 2000;43:292–300. b) Akocak S, Güzel-Akdemir Ö, Kishore Kumar Sanku R, et al. Pyridinium derivatives of 3-aminobenzenesulfonamide are nanomolar-potent inhibitors of tumor-expressed carbonic anhydrase isozymes CA IX and CA XII. Bioorg Chem 2020;103:104204. c) Pastorekova S, Casini A, Scozzafava A, et al. Carbonic anhydrase inhibitors: the first selective, membrane-impermeant inhibitors targeting the tumor-associated isozyme IX. Bioorg Med Chem Lett 2004;14:869–73.
  • a) Supuran CT. Acetazolamide for the treatment of idiopathic intracranial hypertension. Expert Rev Neurother 2015;15:851–6.b) Supuran CT. Applications of carbonic anhydrases inhibitors in renal and central nervous system diseases. Expert Opin Ther Pat 2018;28:713–21.
  • Krasavin M, Kalinin S, Sharonova T, Supuran CT. Inhibitory activity against carbonic anhydrase IX and XII as a candidate selection criterion in the development of new anticancer agents. J Enzyme Inhib Med Chem 2020;35:1555–61.
  • Robertson N, Potter C, Harris AL. Role of carbonic anhydrase IX in human tumor cell growth, survival, and invasion. Cancer Res 2004;64:6160–5.
  • Parkkila S, Rajaniemi H, Parkkila AK, et al. Carbonic anhydrase inhibitor suppresses invasion of renal cancer cells in vitro. Proc Natl Acad Sci U S A 2000;97:2220–4.
  • a) McDonald PC, Chafe SC, Brown WS, et al. Regulation of pH by carbonic anhydrase 9 mediates survival of pancreatic cancer cells with activated KRAS in response to hypoxia. Gastroenterology 2019;157:823–37. b) Chafe SC, McDonald PC, Saberi S, et al. Targeting hypoxia-induced carbonic anhydrase IX enhances immune-checkpoint blockade locally and systemically. Cancer Immunol Res 2019;7:1064–78.
  • a) Peppicelli S, Andreucci E, Ruzzolini J, et al. The carbonic anhydrase IX inhibitor SLC-0111 as emerging agent against the mesenchymal stem cell-derived pro-survival effects on melanoma cells. J Enzyme Inhib Med Chem 2020;35:1185–93. b) Gros SJ, Holland-Cunz SG, Supuran CT, Braissant O. Personalized treatment response assessment for rare childhood tumors using microcalorimetry-exemplified by use of carbonic anhydrase IX and aquaporin 1 inhibitors. Int J Mol Sci 2019;20:4984. c) Petrenko M, Güttler A, Funtan A, et al. Combined 3-O-acetylbetulin treatment and carbonic anhydrase IX inhibition results in additive effects on human breast cancer cells. Chem Biol Interact 2021;333:109326.
  • D’Ascenzio M, Secci D, Carradori S, et al. 1,3-Dipolar cycloaddition, HPLC enantioseparation, and docking studies of saccharin/isoxazole and saccharin/isoxazoline derivatives as selective carbonic anhydrase IX and XII inhibitors. J Med Chem 2020;63:2470–88.