375
Views
44
CrossRef citations to date
0
Altmetric
Review

Carbonic anhydrase inhibitors: an update on experimental agents for the treatment and imaging of hypoxic tumors

Pages 1197-1208 | Received 19 Sep 2021, Accepted 02 Dec 2021, Published online: 13 Dec 2021

References

  • Semenza GL. Intratumoral Hypoxia and mechanisms of immune evasion mediated by Hypoxia-inducible factors. Physiology (Bethesda). 2021;36(2):73–83.
  • Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–275.
  • Choueiri TK, Kaelin WG Jr. Targeting the HIF2-VEGF axis in renal cell carcinoma. Nat Med. 2020;26(10):1519–1530.
  • Pugh CW, Ratcliffe PJ. New horizons in hypoxia signaling pathways. Exp Cell Res. 2017;356:116–121.
  • Wykoff CC, Beasley NJ, Watson PH, et al. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res. 2000;60:7075–7083.
  • Chia SK, Wykoff CC, Watson PH, et al. Prognostic significance of a novel hypoxia-regulated marker, carbonic anhydrase IX, in invasive breast carcinoma. J Clin Oncol. 2001;19:3660–3668.
  • McDonald PC, Swayampakula M, Dedhar S. Coordinated regulation of metabolic transporters and migration/invasion by carbonic anhydrase IX. Metabolites. 2018;8:E20.
  • Chafe SC, McDonald PC, Dedhar S. pH regulators of the tumor microenvironment. A general overview. In: Supuran CT, Carradori S, editors. pH-interfering agents as chemosensitizers in cancer therapy. London (UK): Elsevier; 2021. p. 13–33.
  • Benej M, Svastova E, Banova R, et al. CA IX stabilizes intracellular pH to maintain metabolic reprogramming and proliferation in hypoxia. Front Oncol. 2020;10:1462.
  • Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov. 2011;10:767–777.
  • Supuran CT. Carbonic anhydrase inhibitors as emerging agents for the treatment and imaging of hypoxic tumors. Expert Opin Investig Drugs. 2018;27(12):963–970.
  • Angeli A, Carta F, Nocentini A, et al. Carbonic anhydrase inhibitors targeting metabolism and tumor microenvironment. Metabolites. 2020 14; 10(10): E412.
  • Supuran CT. Carbonic anhydrase inhibition and the management of hypoxic tumors. Metabolites. 2017;7(3):48.
  • Supuran CT. Experimental carbonic anhydrase inhibitors for the treatment of hypoxic tumors. J Exp Pharmacol. 2020;12:603–617.
  • Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov. 2008;7:168–181.
  • Supuran CT. Emerging role of carbonic anhydrase inhibitors. Clin Sci (Lond). 2021;135(10):1233–1249.
  • 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(1):561–580.
  • 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(35):eabj0364.
  • Mboge MY, Combs J, Singh S, et al. Inhibition of carbonic anhydrase using SLC-149: support for a noncatalytic function of CAIX in breast cancer. J Med Chem. 2021;64(3):1713–1724.
  • Supuran CT, Scozzafava A. Benzolamide is not a membrane-impermeant carbonic anhydrase inhibitor. J Enzyme Inhib Med Chem. 2004;19:269–273.
  • Supuran CT. An update on drug interaction considerations in the therapeutic use of carbonic anhydrase inhibitors. Expert Opin Drug Metab Toxicol. 2020;16(4):297–307.
  • Garbati P, Barbieri R, Calderoni M, et al. Efficacy of a three drug-based therapy for neuroblastoma in mice. Int J Mol Sci. 2021;22(13):6753.
  • 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;54:1896–1902.
  • 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.
  • 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 (Camb). 2010;46:8371–8373.
  • 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.
  • Mboge MY, Mahon BP, Lamas N, et al. Structure activity study of carbonic anhydrase IX: selective inhibition with ureido-substituted benzenesulfonamides. Eur J Med Chem. 2017;132:184–191.
  • 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(3):823–837.
  • 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(1):1185–1193.
  • 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(7):1064–1078.
  • Gros SJ, Holland-Cunz SG, Supuran CT, et al. 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(20):4984.
  • Genah S, Angeli A, Supuran CT, et al. Effect of carbonic anhydrase IX inhibitors on human endothelial cell survival. Pharmacol Res. 2020;159:104964.
  • Riemann A, Güttler A, Haupt V, et al. Inhibition of carbonic Anhydrase IX by Ureidosulfonamide inhibitor U104 reduces prostate cancer cell growth, but does not modulate Daunorubicin or Cisplatin cytotoxicity. Oncol Res. 2018;26(2):191–200.
  • Bernardino RL, Dias TR, Moreira BP, et al. Carbonic anhydrases are involved in mitochondrial biogenesis and control the production of lactate by human Sertoli cells. FEBS J. 2019;286(7):1393–1406.
  • 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.
  • 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(7):484–490.
  • Li Z, Jiang L, Chew SH, et al. Carbonic anhydrase 9 confers resistance to ferroptosis/apoptosis in malignant mesothelioma under hypoxia. Redox Biol. 2019;26:101297.
  • Li Z, Jiang L, Toyokuni S. Role of carbonic anhydrases in ferroptosis-resistance. Arch Biochem Biophys. 2020;689:108440.
  • Lee SH, Griffiths JR. How and why are cancers acidic? Carbonic anhydrase IX and the homeostatic control of tumour extracellular pH. Cancers (Basel). 2020;12(6):1616.
  • Morgan PE, Pastoreková S, Stuart-Tilley AK, et al. Interactions of transmembrane carbonic anhydrase, CAIX, with bicarbonate transporters. Am J Physiol Cell Physiol. 2007;293(2):C738–48.
  • Chen Z, Ai L, Mboge MY, et al. UFH-001 cells: a novel triple negative, CAIX-positive, human breast cancer model system. Cancer Biol Ther. 2018;19(7):598–608.
  • Hedlund EE, McDonald PC, Nemirovsky O, et al. Harnessing Induced essentiality: targeting carbonic anhydrase IX and angiogenesis reduces lung metastasis of triple negative breast cancer xenografts. Cancers (Basel). 2019;11(7):1002.
  • Mboge MY, Chen Z, Khokhar D, et al. A non-catalytic function of carbonic anhydrase IX contributes to the glycolytic phenotype and pH regulation in human breast cancer cells. Biochem J. 2019;476(10):1497–1513.
  • 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(11):5591–5600.
  • Bryant JL, Gieling RG, Meredith SL, et al. Novel carbonic anhydrase IX-targeted therapy enhances the anti-tumour effects of cisplatin in small cell lung cancer. Int J Cancer. 2018;142(1):191–201.
  • Xu P, Zhang Y, Ge F, et al. Modulation of tumor microenvironment to enhance radiotherapy efficacy in esophageal squamous cell carcinoma by inhibiting carbonic Anhydrase IX. Front Oncol. 2021;11:637252.
  • 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(14):6328–6338.
  • Akocak S, Lolak N, Bua S, et al. Synthesis and biological evaluation of novel N, N’-diaryl cyanoguanidines acting as potent and selective carbonic anhydrase II inhibitors. Bioorg Chem. 2018;77:245–251.
  • Lolak N, Akocak S, Bua S, et al. Design and synthesis of novel 1,3-diaryltriazene-substituted sulfonamides as potent and selective carbonic anhydrase II inhibitors. Bioorg Chem. 2018;77:542–547.
  • Lomelino CL, Mahon BP, Carta F, et al. Kinetic and X-ray crystallographic investigations on carbonic anhydrase isoforms I, II, IX and XII of a thioureido analog of SLC-0111. Bioorg Med Chem. 2016;24:976–981.
  • Angeli A, Tanini D, Peat TS, et al. Discovery of new selenoureido analogues of 4-(4-fluorophenylureido) benzenesulfonamide as carbonic anhydrase inhibitors ACS. Med Chem Lett. 2017;8:963–968.
  • Eldehna WM, Abo-Ashour MF, Berrino E, et al. SLC-0111 enaminone analogs, 3/4-(3-aryl-3-oxopropenyl) aminobenzenesulfonamides, as novel selective subnanomolar inhibitors of the tumor-associated carbonic anhydrase isoform IX. Bioorg Chem. 2019;83:549–558.
  • Congiu C, Onnis V, Deplano A, et al. Synthesis of sulfonamides incorporating piperazinyl-ureido moieties and their carbonic anhydrase I, II, IX and XII inhibitory activity. Bioorg Med Chem Lett. 2015;25(18):3850–3853.
  • Moi D, Nocentini A, Deplano A, et al. Appliance of the piperidinyl-hydrazidoureido linker to benzenesulfonamide compounds: synthesis, in vitro and in silico evaluation of potent carbonic anhydrase II, IX and XII inhibitors. Bioorg Chem. 2020;98:103728.
  • Abo-Ashour MF, Eldehna WM, Nocentini A, et al. Novel synthesized SLC-0111 thiazole and thiadiazole analogues: determination of their carbonic anhydrase inhibitory activity and molecular modeling studies. Bioorg Chem. 2019;87:794–802.
  • Alkhaldi AAM, Al-Sanea MM, Nocentini A, et al. 3-Methylthiazolo[3,2-a]benzimidazole-Benzenesulfonamide conjugates as novel carbonic Anhydrase inhibitors endowed with anticancer activity: design, synthesis, biological and molecular modeling studies. Eur J Med Chem. 2020;207:112745.
  • Eldehna WM, Fares M, Ceruso M, et al. Amido/ureidosubstituted benzenesulfonamides-isatin conjugates as low nanomolar/subnanomolar inhibitors of the tumor-associated carbonic anhydrase isoform XII. Eur J Med Chem. 2016;110:259–266.
  • Eldehna WM, Abo-Ashour MF, Nocentini A, et al. Enhancement of the tail hydrophobic interactions within the carbonic anhydrase IX active site via structural extension: design and synthesis of novel N-substituted isatins-SLC-0111 hybrids as carbonic anhydrase inhibitors and antitumor agents. Eur J Med Chem. 2019;162:147–160.
  • Eldeeb AH, Abo-Ashour MF, Angeli A, et al. Novel benzenesulfonamides aryl and arylsulfone conjugates adopting tail/dual tail approaches: synthesis, carbonic anhydrase inhibitory activity and molecular modeling studies. Eur J Med Chem. 2021;221:113486.
  • Elbadawi MM, Eldehna WM, Nocentini A, et al. Identification of N-phenyl-2-(phenylsulfonyl)acetamides/propanamides as new SLC-0111 analogues: synthesis and evaluation of the carbonic anhydrase inhibitory activities. Eur J Med Chem. 2021;218:113360.
  • Shaldam M, Eldehna WM, Nocentini A, et al. Development of novel benzofuran-based SLC-0111 analogs as selective cancer-associated carbonic anhydrase isoform IX inhibitors. Eur J Med Chem. 2021;216:113283.
  • Iikuni S, Ono M, Watanabe H, et al. Cancer radiotheranostics targeting carbonic anhydrase-IX with 111In- and 90Y-labeled ureidosulfonamide scaffold for SPECT imaging and radionuclide-based therapy. Theranostics. 2018;8(11):2992–3006.
  • Iikuni S, Watanabe H, Shimizu Y, et al. PET imaging and pharmacological therapy targeting carbonic anhydrase-IX high-expressing tumors using US2 platform based on bivalent ureidosulfonamide. PLoS One. 2020;15(12):e0243327.
  • Iikuni S, Okada Y, Shimizu Y, et al. Modulation of the pharmacokinetics of a Radioligand targeting carbonic Anhydrase-IX with Albumin-Binding Moieties. Mol Pharm. 2021;18(3):966–975.
  • Bua S, Lomelino C, Murray AB, et al. “A sweet combination”: developing Saccharin and Acesulfame K structures for selectively targeting the tumor-associated carbonic Anhydrases IX and XII. J Med Chem. 2020;63(1):321–333.
  • 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(5):2470–2488.
  • 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.
  • Abdelrahman MA, Ibrahim HS, Nocentini A, et al. Novel 3-substituted coumarins as selective human carbonic anhydrase IX and XII inhibitors: synthesis, biological and molecular dynamics analysis. Eur J Med Chem. 2021;209:112897.
  • Dar’in D, Kantin G, Kalinin S, et al. Investigation of 3-sulfamoyl coumarins against cancer-related IX and XII isoforms of human carbonic anhydrase as well as cancer cells leads to the discovery of 2-oxo-2H-benzo[h]chromene-3-sulfonamide - A new caspase-activating proapoptotic agent. Eur J Med Chem. 2021;222:113589.
  • Güttler A, Eiselt Y, Funtan A, et al. Betulin sulfonamides as carbonic anhydrase inhibitors and anticancer agents in breast cancer cells. Int J Mol Sci. 2021;22(16):8808.
  • Baglini E, Ravichandran R, Berrino E, et al. Tetrahydroquinazole-based secondary sulphonamides as carbonic anhydrase inhibitors: synthesis, biological evaluation against isoforms I, II, IV, and IX, and computational studies. J Enzyme Inhib Med Chem. 2021;36(1):1874–1883.
  • Antal I, Koneracka M, Kubovcikova M, et al. Targeting of carbonic anhydrase IX-positive cancer cells by glycine-coated superparamagnetic nanoparticles. Colloids Surf B Biointerfaces. 2021;205:111893.
  • Angeli A, Pinteala M, Maier SS, et al. Tellurides bearing benzensulfonamide as carbonic anhydrase inhibitors with potent antitumor activity. Bioorg Med Chem Lett. 2021;45:128147.
  • Kugler M, Nekvinda J, Holub J, et al. Inhibitors of CA IX enzyme based on polyhedral Boron compounds. Chembiochem. 2021;22(18):2741–2761.
  • Oehler S, Catalano M, Scapozza I, et al. Affinity selections of DNA-encoded chemical libraries on carbonic Anhydrase IX-expressing tumor cells reveal a dependence on ligand valence. Chemistry. 2021 Jun 21;27(35):8985–8993.
  • Cazzamalli S, Dal Corso A, Widmayer F, et al. Chemically defined antibody- and small molecule-drug conjugates for in vivo tumor targeting applications: a comparative analysis. J Am Chem Soc. 2018;140(5):1617–1621.
  • Cazzamalli S, Ziffels B, Widmayer F, et al. Enhanced therapeutic activity of non-internalizing small-molecule-drug conjugates targeting carbonic Anhydrase IX in combination with targeted Interleukin-2. Clin Cancer Res. 2018;24(15):3656–3667.
  • Cazzamalli S, Figueras E, Pethő L, et al. In vivo antitumor activity of a novel acetazolamide-cryptophycin conjugate for the treatment of renal cell carcinomas. ACS Omega. 2018;3(11):14726–14731.
  • Gouyou B, Millul J, Villa A, et al. Sortase-mediated site-specific modification of Interleukin-2 for the generation of a tumor-targeting acetazolamide-cytokine conjugate. ACS Omega. 2020;5(40):26077–26083.
  • Mondal UK, Doroba K, Shabana AM, et al. PEG linker length strongly affects tumor cell killing by PEGylated carbonic anhydrase inhibitors in hypoxic carcinomas expressing carbonic anhydrase IX. Int J Mol Sci. 2021;22(3):1120.
  • Sarnella A, D’Avino G, Hill BS, et al. A novel inhibitor of carbonic anhydrases prevents Hypoxia-induced TNBC cell plasticity. Int J Mol Sci. 2020;21(21):8405.
  • Krasavin M, Kalinin S, Sharonova T, et al. 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(1):1555–1561.
  • Burianova V, Kalinin S, Supuran CT, et al. Radiotracers for positron emission tomography (PET) targeting tumour-associated carbonic anhydrase isoforms. Eur J Med Chem. 2021;213:113046.
  • 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(1):703–711.
  • Sharma V, Kumar R, Angeli A, et al. Tail approach synthesis of novel benzenesulfonamides incorporating 1,3,4-oxadiazole hybrids as potent inhibitor of carbonic anhydrase I, II, IX, and XII isoenzymes. Eur J Med Chem. 2020;193:112219.
  • Ciccone V, Filippelli A, Angeli A, et al. Pharmacological Inhibition of CA-IX impairs tumor cell proliferation, migration and invasiveness. Int J Mol Sci. 2020;21(8):2983.
  • Venkateswaran G, Dedhar S. Interplay of carbonic anhydrase IX with amino acid and acid/base transporters in the hypoxic tumor microenvironment. Front Cell Dev Biol. 2020;8:602668.
  • Mishra CB, Mongre RK, Prakash A, et al. Anti-breast cancer action of carbonic anhydrase IX inhibitor 4-[4-(4-Benzo[1,3]dioxol-5-ylmethyl-piperazin-1-yl)-benzylidene-hydrazinocarbonyl]-benzenesulfonamide (BSM-0004): in vitro and in vivo studies. J Enzyme Inhib Med Chem. 2021;36(1):954–963.
  • Temiz E, Koyuncu I, Durgun M, et al. Inhibition of carbonic anhydrase IX promotes apoptosis through Intracellular pH level alterations in cervical cancer cells. Int J Mol Sci. 2021;22(11):6098.
  • Yamali C, Inci Gul H, Ozli G, et al. Exploring of tumor-associated carbonic anhydrase isoenzyme IX and XII inhibitory effects and cytotoxicities of the novel N-aryl-1-(4-sulfamoylphenyl)-5-(thiophen-2-yl)-1H-pyrazole-3-carboxamides. Bioorg Chem. 2021;115:105194.
  • Elimam DM, Elgazar AA, Bonardi A, et al. Natural inspired piperine-based sulfonamides and carboxylic acids as carbonic anhydrase inhibitors: design, synthesis and biological evaluation. Eur J Med Chem. 2021;225:113800.
  • Millul J, Krudewig C, Zana A, et al. Immunotherapy with Immunocytokines and PD-1 blockade enhances the anticancer activity of small molecule-drug conjugates targeting carbonic anhydrase IX. Mol Cancer Ther. 2021;20(3):512–522.
  • Supuran CT. Multitargeting approaches involving carbonic anhydrase inhibitors: hybrid drugs against a variety of disorders. J Enzyme Inhib Med Chem. 2021;36(1):1702–1714.
  • 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.
  • Marks IS, Gardeen SS, Kurdziel SJ, et al. Development of a small molecule Tubulysin B conjugate for treatment of carbonic anhydrase IX receptor expressing cancers. Mol Pharm. 2018;15:2289–2296.
  • Buller F, Steiner M, Frey K, et al. Selection of carbonic anhydrase IX inhibitors from one million DNA-encoded compounds. ACS Chem Biol. 2011;6:336–344.
  • An R, Lin B, Zhao S, et al. Discovery of novel artemisinin-sulfonamide hybrids as potential carbonic anhydrase IX inhibitors with improved antiproliferative activities. Bioorg Chem. 2020;104:104347.
  • Yu H, Hou Z, Tian Y, et al. Design, synthesis, cytotoxicity and mechanism of novel dihydroartemisinin-coumarin hybrids as potential anti-cancer agents. Eur J Med Chem. 2018 May 10;151:434–449.
  • Tian Y, Liang Z, Xu H, et al. Design, synthesis and cytotoxicity of novel Dihydroartemisinin-Coumarin hybrids via click chemistry. Molecules. 2016;21(6):758.
  • Zhang B, Liu Z, Xia S, et al. Design, synthesis and biological evaluation of sulfamoylphenyl-quinazoline derivatives as potential EGFR/CAIX dual inhibitors. Eur J Med Chem. 2021;216:113300.
  • Elzahhar PA, Abd El Wahab SM, Elagawany M, et al. Expanding the anticancer potential of 1,2,3-triazoles via simultaneously targeting Cyclooxygenase-2, 15-lipoxygenase and tumor-associated carbonic anhydrases. Eur J Med Chem. 2020;200:112439.
  • Berrino E, Angeli A, Zhdanov DD, et al. Azidothymidine “Clicked” into 1,2,3-Triazoles: first report on carbonic Anhydrase-Telomerase dual-hybrid inhibitors. J Med Chem. 2020;63:7392–7409.
  • Teodori E, Braconi L, Bua S, et al. Dual P-Glycoprotein and CA XII inhibitors: a new strategy to reverse the P-gp mediated multidrug resistance (MDR) in cancer cells. Molecules. 2020;25:1748.
  • Krasavin M, Sharonova T, Sharoyko V, et al. Combining carbonic anhydrase and thioredoxin reductase inhibitory motifs within a single molecule dramatically increases its cytotoxicity. J Enzyme Inhib Med Chem. 2020;35(1):665–671.
  • Ceni C, Catarzi D, Varano F, et al. Discovery of first-in-class multi-target adenosine A2A receptor antagonists-carbonic anhydrase IX and XII inhibitors. 8-Amino-6-aryl-2-phenyl-1,2,4-triazolo [4,3-a]pyrazin-3-one derivatives as new potential antitumor agents. Eur J Med Chem. 2020;201:112478.
  • Lee SY, Namasivayam V, Boshta NM, et al. Discovery of potent nucleotide pyrophosphatase/phosphodiesterase3 (NPP3) inhibitors with ancillary carbonic anhydrase inhibition for cancer (immuno)therapy. RSC Med Chem. 2021;12(7):1187–1206.
  • Petreni A, Bonardi A, Lomelino C, et al. Inclusion of a 5-fluorouracil moiety in nitrogenous bases derivatives as human carbonic anhydrase IX and XII inhibitors produced a targeted action against MDA-MB-231 and T47D breast cancer cells. Eur J Med Chem. 2020;190:112112.
  • Mujumdar P, Kopecka J, Bua S, et al. Carbonic anhydrase XII inhibitors overcome temozolomide resistance in glioblastoma. J Med Chem. 2019;62(8):4174–4192.
  • Battke C, Kremmer E, Mysliwietz J, et al. Generation and characterization of the first inhibitory antibody targeting tumour-associated carbonic anhydrase XII. Cancer Immunol Immunother. 2011;60(5):649–658.
  • Alterio V, Kellner M, Esposito D, et al. Biochemical and structural insights into carbonic anhydrase XII/Fab6A10 complex. J Mol Biol. 2019;431(24):4910–4921.
  • Gondi G, Mysliwietz J, Hulikova A, et al. Antitumor efficacy of a monoclonal antibody that inhibits the activity of cancer-associated carbonic anhydrase XII. Cancer Res. 2013;73(21):6494–6503.
  • Fiedler L, Kellner M, Oos R, et al. Fully automated production and characterization of 64 Cu and proof-of-principle small-animal PET imaging using 64 Cu-labelled CA XII targeting 6A10 Fab. ChemMedChem. 2018;13(12):1230–1237.
  • Fiedler L, Kellner M, Gosewisch A, et al. Evaluation of 177Lu[Lu]-CHX-A″-DTPA-6A10 Fab as a radioimmunotherapy agent targeting carbonic anhydrase XII. Nucl Med Biol. 2018;60:55–62.
  • Von Neubeck B, Gondi G, Riganti C, et al. An inhibitory antibody targeting carbonic anhydrase XII abrogates chemoresistance and significantly reduces lung metastases in an orthotopic breast cancer model in vivo. Int J Cancer. 2018;143(8):2065–2075.
  • Li G, Chen TW, Nickel AC, et al. Carbonic anhydrase XII is a clinically significant, molecular tumor-subtype specific therapeutic target in glioma with the potential to combat invasion of brain tumor cells. Onco Targets Ther. 2021;14:1707–1718.
  • Zatovicova M, Jelenska L, Hulikova A, et al. Carbonic anhydrase IX as an anticancer therapy target: preclinical evaluation of internalizing monoclonal antibody directed to catalytic domain. Curr Pharm Des. 2010;16(29):3255–3263.
  • Mishra CB, Tiwari M, Supuran CT. Progress in the development of human carbonic anhydrase inhibitors and their pharmacological applications: where are we today? Med Res Rev. 2020;40:2485–2565.
  • Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–1072.
  • 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;38:1799–1836.
  • Zheng C, Song Q, Zhao H, et al. A nanoplatform to boost multi-phases of cancer-immunity-cycle for enhancing immunotherapy. J Control Release. 2021;339:403–415.

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.