We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Challenging breast cancer through novel sulfonamide–pyridine hybrids: design, synthesis, carbonic anhydrase IX inhibition and induction of apoptosis

    Nashwa H Zaher

    Drug Radiation Research, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt

    ,
    Reham MM Elhazek

    Drug Radiation Research, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt

    ,
    Ahmed E Gouda

    Pharmaceutical Research Department, Nawah Scientific, Cairo, Egypt

    ,
    Amira Khalil

    *Author for correspondence:

    E-mail Address: Amira.Khalil@bue.edu.eg

    Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt

    The Center for Drug Research & Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt

    &
    Marwa G Elgazzar

    Drug Radiation Research, National Center for Radiation Research & Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt

    Published Online:https://doi.org/10.4155/fmc-2022-0197

    Background: Among the important key modulators of the tumor microenvironment and hypoxia is a family of enzymes named carbonic anhydrases. Herein, 11 novel sulfonamide–pyridine hybrids (2–12) were designed, synthesized and biologically evaluated for their potential use in targeting breast cancer. Methods & results: The para chloro derivative 7 reported the highest cytotoxic activity against the three breast cancer cell lines used. In addition, compound 7 was found to induce cell cycle arrest and autophagy as well as delaying wound healing. The IC50 of compound 7 against carbonic anhydrase IX was 253 ± 12 nM using dorzolamide HCl as control. Conclusion: This study encourages us to expand the designed library, where more sulfonamide derivatives would be synthesized and studied for their structure–activity relationships.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Rashid HU, Xu YM, Muhammad Y, Wang LS, Jiang J. Research advances on anticancer activities of matrine and its derivatives: an updated overview. Eur. J. Med. Chem. 161, 205–238 (2019).
    • 2. Ahmadi R, Ebrahimzadeh MA. Resveratrol – a comprehensive review of recent advances in anticancer drug design and development. Eur. J. Med. Chem. 200, 112356 (2020). • Highlights the major important advances in the field of drug discovery for anticancer agents.
    • 3. Liang T, Sun X, Li W, Hou G, Gao F. 1,2,3-Triazole-containing compounds as anti-lung cancer agents: current developments, mechanisms of action, and structure–activity relationship. Front. Pharmacol. 12, 661173 (2021).
    • 4. Jarak I, Varela CL, Da Silva ET, Roleira FFM, Veiga F, Figueiras A. Pluronic-based nanovehicles: recent advances in anticancer therapeutic applications. Eur. J. Med. Chem. 206, 112526 (2020).
    • 5. Cui WQ, Aouidate A, Wang SG, Yu QLY, Li YH, Yuan SG. Discovering anti-cancer drugs via computational methods. Front. Pharmacol. 11, 733 (2020).
    • 6. Anderson NM, Simon MC. The tumor microenvironment. Curr. Biol. 30(16), R921–R925 (2020).
    • 7. Emami Nejad A, Najafgholian S, Rostami A et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int. 21(1), 62 (2021).
    • 8. Gillies RJ, Brown JS, Anderson ARA, Gatenby RA. Eco-evolutionary causes and consequences of temporal changes in intratumoural blood flow. Nat. Rev. Cancer 18(9), 576–585 (2018).
    • 9. Pastorekova S, Gillies RJ. The role of carbonic anhydrase IX in cancer development: links to hypoxia, acidosis, and beyond. Cancer Metastasis Rev. 38(1-2), 65–77 (2019).
    • 10. Nejad AE, Najafgholian S, Rostami A et al. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int. 21(1), 62 (2021).
    • 11. Shi R, Liao C, Zhang Q. Hypoxia-driven effects in cancer: characterization, mechanisms, and therapeutic implications. Cells 10(3), 678 (2021).
    • 12. Ratcliffe PJ. Oxygen sensing and hypoxia signalling pathways in animals: the implications of physiology for cancer. J. Physiol. 591(8), 2027–2042 (2013).
    • 13. Kim LC, Simon MC. Hypoxia-inducible factors in cancer. Cancer Res. 82(2), 195–196 (2022).
    • 14. Guarnaccia L, Navone SE, Trombetta E et al. Angiogenesis in human brain tumors: screening of drug response through a patient-specific cell platform for personalized therapy. Sci. Rep. 8(1), 8748 (2018).
    • 15. Ahn GO, Seita J, Hong BJ et al. Transcriptional activation of hypoxia-inducible factor-1 (HIF-1) in myeloid cells promotes angiogenesis through VEGF and S100A8. Proc. Natl Acad. Sci. USA 111(7), 2698–2703 (2014).
    • 16. Pastorek J, Pastorekova S. Hypoxia-induced carbonic anhydrase IX as a target for cancer therapy: from biology to clinical use. Semin. Cancer Biol. 31, 52–64 (2015).
    • 17. Pastorek J, Pastorekova S, Callebaut I et al. Cloning and characterization of MN, a human tumor-associated protein with a domain homologous to carbonic anhydrase and a putative helix-loop-helix DNA binding segment. Oncogene 9(10), 2877–2888 (1994).
    • 18. Kumar S, Rulhania S, Jaswal S, Monga V. Recent advances in the medicinal chemistry of carbonic anhydrase inhibitors. Eur. J. Med. Chem. 209(1), 112923 (2021).
    • 19. Parkkila S, Parkkila AK, Saarnio J et al. Expression of the membrane-associated carbonic anhydrase isozyme XII in the human kidney and renal tumors. J. Histochem. Cytochem. 48(12), 1601–1608 (2000).
    • 20. Haapasalo J, Hilvo M, Nordfors K et al. Identification of an alternatively spliced isoform of carbonic anhydrase XII in diffusely infiltrating astrocytic gliomas. Neuro Oncol. 10(2), 131–138 (2008).
    • 21. Korhonen K, Parkkila AK, Helen P et al. Carbonic anhydrases in meningiomas: association of endothelial carbonic anhydrase II with aggressive tumor features. J. Neurosurg. 111(3), 472–477 (2009).
    • 22. Lou YM, Mcdonald PC, Oloumi A et al. Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Res. 71(9), 3364–3376 (2011).
    • 23. Sung H, Ferlay J, Siegel RL et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71(3), 209–249 (2021).
    • 24. Nguyen QH, Pervolarakis N, Blake K et al. Profiling human breast epithelial cells using single cell RNA sequencing identifies cell diversity. Nat. Commun. 9(1), 2028 (2018). • Properties of cancer cells and illustration of cell heterogenity.
    • 25. Kogel D, Linder B, Brunschweiger A, Chines S, Behl C. At the crossroads of apoptosis and autophagy: multiple roles of the co-chaperone BAG3 in stress and therapy resistance of cancer. Cells 9(3), 574 ((.2020). •• The role of apoptosis and autophagy in cancer cell death.
    • 26. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 144(5), 646–674 (2011). • Very important review covering many interesting points related to our current work.
    • 27. King EK, Losier TT, Russell CR. Regulation of autophagy enzymes by nutrient signaling. Trends Biochem. Sci. 46(8), 687–700 (2021). • Highlights the importance of autophagy.
    • 28. Di Bartolomeo S, Latella L, Zarbalis K, Di Sano F. Editorial: autophagy in mammalian development and differentiation. Front. Cell Dev. Biol. 9, 722821 (2021).
    • 29. Fang YY, Tan J, Zhang Q. Signaling pathways and mechanisms of hypoxia-induced autophagy in the animal cells. Cell Biol. Int. 39(8), 891–898 (2015).
    • 30. Lim KH, Staudt LM. Toll-like receptor signaling. Cold Spring Harb. Perspect. Biol. 5(1), a011247 (2013).
    • 31. Rosenfeldt MT, Ryan KM. The multiple roles of autophagy in cancer. Carcinogenesis 32(7), 955–963 (2011).
    • 32. Ghorab MM, Heiba HI, Khalil AI, El Ella DaA, Noaman E. Computer-based ligand design and synthesis of some new sulfonamides bearing pyrrole or pyrrolopyrimidine moieties having potential antitumor and radioprotective activities. Phosphorus Sulfur Rel. Elem. 183(1), 90–104 (2008).
    • 33. Muller-Schiffmann A, Marz-Berberich J, Andreyeva A et al. Combining independent drug classes into superior, synergistically acting hybrid molecules. Angew. Chem. Int. Edit. Engl. 49(46), 8743–8746 (2010).
    • 34. Ghorab MM, El-Gazzar MG, Alsaid MS. Synthesis and anti-breast cancer evaluation of novel N-(guanidinyl)benzenesulfonamides. Int. J. Mol. Sci. 15(4), 5582–5595 (2014). •• Includes important chemical and biological details related to our study.
    • 35. 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. 110, 259–266 (2016). •• Important article relating to sulfonamides and carbonic anydrase inhibition.
    • 36. Ghorab MM, Alsaid MS, Al-Ansary GH, Abdel-Latif GA, Abou El Ella DA. Analogue based drug design, synthesis, molecular docking and anticancer evaluation of novel chromene sulfonamide hybrids as aromatase inhibitors and apoptosis enhancers. Eur. J. Med. Chem. 124, 946–958 (2016).
    • 37. Eldehna WM, Abo-Ashour MF, Nocentini A et al. Novel 4/3-((4-oxo-5-(2-oxoindolin-3-ylidene)thiazolidin-2-ylidene) amino) benzenesulfonamides: synthesis, carbonic anhydrase inhibitory activity, anticancer activity and molecular modelling studies. Eur. J. Med. Chem. 139, 250–262 (2017).
    • 38. Karali N, Akdemir A, Goktas F et al. Novel sulfonamide-containing 2-indolinones that selectively inhibit tumor-associated alpha carbonic anhydrases. Bioorg. Med. Chem. 25(14), 3714–3718 (2017).
    • 39. Supuran CT. Advances in structure-based drug discovery of carbonic anhydrase inhibitors. Expert Opin. Drug Discov. 12(1), 61–88 (2017).
    • 40. Eldehna WM, Nocentini A, Al-Rashood ST et al. Tumor-associated carbonic anhydrase isoform IX and XII inhibitory properties of certain isatin-bearing sulfonamides endowed with in vitro antitumor activity towards colon cancer. Bioorg. Chem. 81, 425–432 (2018).
    • 41. Gul HI, Yamali C, Sakagami H et al. New anticancer drug candidates sulfonamides as selective hCA IX or hCA XII inhibitors. Bioorg. Chem. 77, 411–419 (2018).
    • 42. Nocentini A, Trallori E, Singh S et al. 4-Hydroxy-3-nitro-5-ureido-benzenesulfonamides selectively target the tumor-associated carbonic anhydrase isoforms IX and XII showing hypoxia-enhanced antiproliferative profiles. J. Med. Chem. 61(23), 10860–10874 (2018).
    • 43. Peerzada MN, Khan P, Ahmad K, Hassan MI, Azam A. Synthesis, characterization and biological evaluation of tertiary sulfonamide derivatives of pyridyl–indole based heteroaryl chalcone as potential carbonic anhydrase IX inhibitors and anticancer agents. Eur. J. Med. Chem. 155, 13–23 (2018).
    • 44. Cecchi A, Winum JY, Innocenti A et al. Carbonic anhydrase inhibitors: synthesis and inhibition of cytosolic/tumor-associated carbonic anhydrase isozymes I, II, and IX with sulfonamides derived from 4-isothiocyanato-benzolamide. Bioorg. Med. Chem. Lett. 14(23), 5775–5780 (2004).
    • 45. Garaj V, Puccetti L, Fasolis G et al. Carbonic anhydrase inhibitors: synthesis and inhibition of cytosolic/tumor-associated carbonic anhydrase isozymes I, II, and IX with sulfonamides incorporating 1,2,4-triazine moieties. Bioorg. Med. Chem. Lett. 14(21), 5427–5433 (2004).
    • 46. Aday B, Ulus R, Tanc M, Kaya M, Supuran CT. Synthesis of novel 5-amino-1,3,4-thiadiazole-2-sulfonamide containing acridine sulfonamide/carboxamide compounds and investigation of their inhibition effects on human carbonic anhydrase I, II, IV and VII. Bioorg. Chem. 77, 101–105 (2018).
    • 47. Koch G. Medicinal chemistry: editorial. CHIMIA 71(10), 643 (2017).
    • 48. 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. 43(7), 484–490 (2020).
    • 49. Gieling RG, Babur M, Mamnani L et al. Antimetastatic effect of sulfamate carbonic anhydrase IX inhibitors in breast carcinoma xenografts. J. Med. Chem. 55(11), 5591–5600 (2012).
    • 50. Williams KJ, Gieling RG. Preclinical evaluation of ureidosulfamate carbonic anhydrase IX/XII inhibitors in the treatment of cancers. Int. J. Mol. Sci. 20(23), 6080 (2019).
    • 51. 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. 132, 184–191 (2017).
    • 52. Mboge MY, Chen Z, Wolff A et al. Selective inhibition of carbonic anhydrase IX over carbonic anhydrase XII in breast cancer cells using benzene sulfonamides: disconnect between activity and growth inhibition. PLOS ONE 13(11), e0207417 (2018).
    • 53. Bailón-Moscoso N, Romero-Benavides JC, Ostrosky-Wegman P. Development of anticancer drugs based on the hallmarks of tumor cells. Tumour Biol. 35(5), 3981–3995 (2014).
    • 54. Lovitt CJ, Shelper TB, Avery VM. Doxorubicin resistance in breast cancer cells is mediated by extracellular matrix proteins. BMC Cancer 18(1), 41 (2018).
    • 55. Viegas Junior C, Danuello A, Bolzani V, Barreiro E, Fraga C. Molecular hybridization: a useful tool in the design of new drug prototypes. Curr. Med. Chem. 14, 1829–1852 (2007).
    • 56. Narendar P, Parthiban J, Anbalagan N, Gunasekaran V, Thomas Leonard J. Pharmacological evaluation of some new 2-substituted pyridine derivatives. Biol. Pharm. Bull. 26(2), 182–187 (2003).
    • 57. Menegatti R, Silva GMS, Zapata-Sudo G et al. Design, synthesis, and pharmacological evaluation of new neuroactive pyrazolo[3,4-b]pyrrolo[3,4-d]pyridine derivatives with in vivo hypnotic and analgesic profile. Bioorg. Med. Chem. 14(3), 632–640 (2006).
    • 58. El-Naggar M, Almahli H, Ibrahim HS, Eldehna WM, Abdel-Aziz HA. Pyridine-ureas as potential anticancer agents: synthesis and in vitro biological evaluation. Molecules 23(6), 1459 (2018).
    • 59. Albratty M, Alhazmi HA. Novel pyridine and pyrimidine derivatives as promising anticancer agents: a review. Arabian J. Chem. 15(6), 103846 (2022).
    • 60. 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. 35(1), 1555–1561 (2020).
    • 61. Deka S, Mohan S, Saravanan J et al. Syntheses, characterization and in-vitro anti-Infammatory activity of some novel thiophenes. Maced. J. Med. Sci. 5, 159–163 (2012).
    • 62. Ragab FA, Heiba HI, El-Gazzar MG, Abou-Seri SM, El-Sabbagh WA, El-Hazek RM. Synthesis of novel thiadiazole derivatives as selective COX-2 inhibitors. Med. Chem. Commun. 7(12), 2309–2327 (2016).
    • 63. Berman HM, Westbrook J, Feng Z et al. The Protein Data Bank. Nucleic Acids Res. 28(1), 235–242 (2000).
    • 64. Zakšauskas A, Čapkauskaitė E, Paketurytė-Latvė V et al. Methyl 2-halo-4-substituted-5-sulfamoyl-benzoates as high affinity and selective inhibitors of carbonic anhydrase IX. Int. J. Mol. Sci. 23(1), 130 (2022).
    • 65. Morris GM, Huey R, Lindstrom W et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J. Comput. Chem. 30(16), 2785–2791 (2009).
    • 66. Santos-Martins D, Forli S, Ramos MJ, Olson AJ. AutoDock4(Zn): an improved AutoDock force field for small-molecule docking to zinc metalloproteins. J. Chem. Inf. Model. 54(8), 2371–2379 (2014).
    • 67. O'boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open Babel: an open chemical toolbox. J. Cheminform. 3, 33 (2011).
    • 68. Halgren TA. MMFF VI. MMFF94s option for energy minimization studies. J. Comput. Chem. 20(7), 720–729 (1999).
    • 69. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31(2), 455–461 (2010).
    • 70. Pettersen EF, Goddard TD, Huang CC et al. UCSF Chimera – a visualization system for exploratory research and analysis. J. Comput. Chem. 25(13), 1605–1612 (2004).
    • 71. Jacobs GP. A review of the effects of gamma radiation on pharmaceutical materials. J. Biomater. Appl. 10(1), 59–96 (1995).
    • 72. El-Gaby MSA, Abdel-Gawad SM, Ghorab MM, Heiba HI, Aly HM. Synthesis and biological activity of some novel thieno[2,3-b]quinoline, quinolino[3′,2′:4,5] thieno[3,2-d]pyrimidine and pyrido[2′,3′:4,5] thieno[2,3-b]quinoline derivatives. Phosphorus Sulfur Silicon Relat. Elem. 181(2), 279–297 (2006).
    • 73. Singh BK, Parwate DV, Das Sarma IB, Shukla SK. Study on gamma and electron beam sterilization of third generation cephalosporins cefdinir and cefixime in solid state. Radiat. Phys. Chem. 79(10), 1079–1087 (2010).
    • 74. Crucq A-S, Deridder V, Tilquin B. Radiostability of pharmaceuticals under different irradiation conditions. Radiat. Phys. Chem. 72(2), 355–361 (2005).
    • 75. Zalewski P, Skibiński R, Szymanowska-Powałowska D, Piotrowska H, Bednarski W, Cielecka-Piontek J. Radiolytic studies of cefozopran hydrochloride in the solid state. Electron. J. Biotechnol. 25, 28–32 (2017).
    • 76. Huey R, Morris GM, Olson AJ, Goodsell DS. A semiempirical free energy force field with charge-based desolvation. J. Comput. Chem. 28(6), 1145–1152 (2007).
    • 77. Bell EW, Zhang Y. DockRMSD: an open-source tool for atom mapping and RMSD calculation of symmetric molecules through graph isomorphism. J. Cheminform. 11(1), 40 (2019).
    • 78. Skehan P, Storeng R, Scudiero D et al. New colorimetric cytotoxicity assay for anticancer-drug screening. J. Natl Cancer Inst. 82(13), 1107–1112 (1990).
    • 79. Darzynkiewicz Z, Bedner E, Smolewski P. Flow cytometry in analysis of cell cycle and apoptosis. Semin. Hematol. 38(2), 179–193 (2001).
    • 80. Fathy U, Abd El Salam HA, Fayed EA, Elgamal AM, Gouda A. Facile synthesis and in vitro anticancer evaluation of a new series of tetrahydroquinoline. Heliyon 7(10), e08117 (2021).
    • 81. Bashmail HA, Alamoudi AA, Noorwali A, Hegazy GA, Ajabnoor GM, Al-Abd AM. Thymoquinone enhances paclitaxel anti-breast cancer activity via inhibiting tumor-associated stem cells despite apparent mathematical antagonism. Molecules 25(2), 426 (2020).
    • 82. Yousefi S, Simon HU. Autophagy in cancer and chemotherapy. Results Probl. Cell Differ. 49, 183–190 (2009).
    • 83. Zeng F, Ju RJ, Liu L, Xie HJ, Mu LM, Lu WL. Efficacy in treating lung metastasis of invasive breast cancer with functional vincristine plus dasatinib liposomes. Pharmacology 101(1-2), 43–53 (2018).
    • 84. Supuran CT. Indisulam: an anticancer sulfonamide in clinical development. Expert Opin. Investig. Drugs 12(2), 283–287 (2003).
    • 85. Ruzzolini J, Laurenzana A, Andreucci E et al. A potentiated cooperation of carbonic anhydrase IX and histone deacetylase inhibitors against cancer. J. Enzyme Inhib. Med. Chem. 35(1), 391–397 (2020).
    • 86. 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. 22(11), 6098 (2021).
    • 87. Cianchi F, Vinci MC, Supuran CT et al. Selective inhibition of carbonic anhydrase IX decreases cell proliferation and induces ceramide-mediated apoptosis in human cancer cells. J. Pharmacol. Exp. Ther. 334(3), 710–719 (2010).
    • 88. Koyuncu I, Gonel A, Kocyigit A, Temiz E, Durgun M, Supuran CT. Selective inhibition of carbonic anhydrase-IX by sulphonamide derivatives induces pH and reactive oxygen species-mediated apoptosis in cervical cancer HeLa cells. J. Enzyme Inhib. Med. Chem. 33(1), 1137–1149 (2018).
    • 89. Mohammadpour R, Safarian S, Ejeian F, Sheikholya-Lavasani Z, Abdolmohammadi MH, Sheinabi N. Acetazolamide triggers death inducing autophagy in T-47D breast cancer cells. Cell Biol. Int. 38(2), 228–238 (2014).
    • 90. Ward C, Meehan J, Mullen P et al. Evaluation of carbonic anhydrase IX as a therapeutic target for inhibition of breast cancer invasion and metastasis using a series of in vitro breast cancer models. Oncotarget 6(28), 24856–24870 (2015).