Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter February 2, 2022

Anticancer properties of arylchromenes and arylchromans: an overview

  • Yuri de Freitas Rego , Nathália Evelyn Morais Costa , Rodrigo Martins de Lacerda , Angélica Faleiros da Silva Maia , Cleiton Moreira da Silva ORCID logo and Ângelo de Fátima ORCID logo EMAIL logo
From the journal Physical Sciences Reviews

Abstract

Cancers are a set of pathologies originated by cells that have the ability to divide and multiply uncontrollably, associated with the capacity to invade and colonize adjacent tissues. Chemotherapy is one of the main approaches of treatment for cancer patients. Despite of the numerous antineoplastic drugs available, cancer cannot be cured; particularly at the late stages deprived of any side effect. Arylchromenes and arylchromans are a group of small molecules, of natural or synthetic origin, of great interest as prototypes for the drug development, especially against cancer. In this chapter, we will present the antineoplastic activity studies of the most promising examples of these arylchromenes and arylchroman derivatives.


Corresponding author: Ângelo de Fátima, Departamento de Química, Grupo de Estudos em Química Orgânica e Biológica (GEQOB), Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil, E-mail:

Funding source: Pró-Reitoria de Pesquisa da Universidade Federal de Minas Gerais

Acknowledgments

The authors thank the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Pró-Reitoria de Pesquisa da Universidade Federal de Minas Gerais (PRPq-UFMG) for their support. ADF is supported by a CNPq research fellowship.

  1. Author contribution: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Conflict of interest statement: The authors declare no conflicts of interest regarding this article.

References

1. Alberts, B, Johnson, A, Lewis, J, Morgan, D, Raff, M, Roberts, K, et al.. Molecular Biology of the Cell, 6th ed. New York: Garland Science Taylor & Francis Group; 2015.Search in Google Scholar

2. Brooker, RJ. Genetics: Analysis & Principles, 6th ed. New York: McGraw-Hill Education; 2018.Search in Google Scholar

3. Hanahan, D, Weinberg, RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–74, https://doi.org/10.1016/j.cell.2011.02.013.Search in Google Scholar PubMed

4. Lodish, H, Berk, A, Kaiser, CA, Krieger, M, Bretscher, A, Ploegh, H, et al.. Molecular Cell Biology, 8th ed. New York: Macmillan Learning; 2018.Search in Google Scholar

5. World Health Organization. Who Report on Cancer: Setting Priorities. Geneva: Investing Wisely and Providing Care for All; 2020.Search in Google Scholar

6. Gilman, AG, Brunton, LL. Goodman and Gilman’s The Pharmacological Basis of Therapeutics, Brunton, LL, Hilal-Dandan, R, Knollmann, BC, editors, 13th ed. New York: McGraw-Hill Education; 2017.Search in Google Scholar

7. Holohan, C, Van Schaeybroeck, S, Longley, DB, Johnston, PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013;13:714–26, https://doi.org/10.1038/nrc3599.Search in Google Scholar PubMed

8. Patil, SA, Patil, R, Pfeffer, LM, Miller, DD. Chromenes: potential new chemotherapeutic agents for cancer. Future Med Chem 2013;5:1647–60, https://doi.org/10.4155/fmc.13.126.Search in Google Scholar PubMed

9. Raj, V, Lee, J. 2H/4H-Chromenes—a versatile biologically attractive scaffold. Front Chem 2020;8:1–23, https://doi.org/10.3389/fchem.2020.00623.Search in Google Scholar PubMed PubMed Central

10. Boulmokh, Y, Belguidoum, K, Meddour, F, Amira-Guebailia, H. Investigation of antioxidant activity of epigallocatechin gallate and epicatechin as compared to resveratrol and ascorbic acid: experimental and theoretical insights. Struct Chem 2021;32:1907–23, https://doi.org/10.1007/s11224-021-01763-5.Search in Google Scholar

11. Knidel, C, Pereira, MF, Barcelos, DHF, de Oliveira Gomes, DC, Guimarães, MCC, Schuenck, RP. Epigallocatechin gallate has antibacterial and antibiofilm activity in methicillin resistant and susceptible Staphylococcus aureus of different lineages in non-cytotoxic concentrations. Nat Prod Res 2019;10:1–5, https://doi.org/10.1080/14786419.2019.1698575.Search in Google Scholar PubMed

12. Rahmani-Nezhad, S, Safavi, M, Pordeli, M, Ardestani, SK, Khosravani, L, Pourshojaei, Y, et al.. Synthesis, in vitro cytotoxicity and apoptosis inducing study of 2-aryl-3-nitro-2 H -chromene derivatives as potent anti-breast cancer agents. Eur J Med Chem 2014;86:562–9, https://doi.org/10.1016/j.ejmech.2014.09.017.Search in Google Scholar PubMed

13. Ying, J, Yang, W, Xie, CY, Ni, QC, Pan, XD, Dong, JH, et al.. Induction of caspase-3-dependent apoptosis in human leukemia hl-60 cells by δ-elemene. Yakugaku Zasshi 2011;131:1383–94, https://doi.org/10.1248/yakushi.131.1383.Search in Google Scholar PubMed

14. Sun, Q, Li, FF, Wang, D, Wu, J, Yao, GD, Li, X, et al.. Flavans with cytotoxic activity from the stem and root bark of: Daphne giraldii. RSC Adv 2016;6:55919–29, https://doi.org/10.1039/c6ra08537g.Search in Google Scholar

15. Afsar, T, Trembley, JH, Salomon, CE, Razak, S, Khan, MR, Ahmed, K. Growth inhibition and apoptosis in cancer cells induced by polyphenolic compounds of Acacia hydaspica: involvement of multiple signal transduction pathways. Sci Rep 2016;6:1–12, https://doi.org/10.1038/srep23077.Search in Google Scholar PubMed PubMed Central

16. Liu, J, Mei, WL, Wu, J, Zhao, YX, Peng, M, Dai, HF. A new cytotoxic homoisoflavonoid from Dracaena cambodiana. J Asian Nat Prod Res 2009;11:192–5, https://doi.org/10.1080/10286020802674962.Search in Google Scholar PubMed

17. Hu, J, Zhou, D, Chen, Y. Preparation and antioxidant activity of green tea extract enriched in epigallocatechin (EGC) and epigallocatechin gallate (EGCG). J Agric Food Chem 2009;57:1349–53, https://doi.org/10.1021/jf803143n.Search in Google Scholar PubMed

18. Vergote, D, Cren-Olivé, C, Chopin, V, Toillon, RA, Rolando, C, Hondermarck, H, et al.. (-)-Epigallocatechin (EGC) of green tea induces apoptosis of human breast cancer cells but not of their normal counterparts. Breast Cancer Res Treat 2002;76:195–201, https://doi.org/10.1023/a:1020833410523.10.1023/A:1020833410523Search in Google Scholar PubMed

19. Kinjo, J, Nagao, T, Tanaka, T, Nonaka, GI, Okawa, M, Nohara, T, et al.. Activity-guided fractionation of green tea extract with antiproliferative activity against human stomach cancer cells. Biol Pharm Bull 2002;25:1238–40, https://doi.org/10.1248/bpb.25.1238.Search in Google Scholar PubMed

20. Sharifi-rad, M, Redaelli, M, Zorzan, M, Cho, WC, Sharifi-rad, J. Preclinical activities of epigallocatechin gallate in signaling pathways in cancer. Molecules 2020;25:1–29, https://doi.org/10.3390/molecules25030467.Search in Google Scholar PubMed PubMed Central

21. Yang, LL, Yen, KY, Kiso, Y, Hikino, H. Antihepatotoxic actions of Formosan plant drugs. J Ethnopharmacol 1987;19:103–10, https://doi.org/10.1016/0378-8741(87)90142-5.Search in Google Scholar PubMed

22. Kim, YK, Kim, YS, Choi, SU, Ryu, SY. Isolation of flavonol rhamnosides from Loranthus tanakae and cytotoxic effect of them on human tumor cell lines. Arch Pharm Res (Seoul) 2004;27:44–7, https://doi.org/10.1007/bf02980044.Search in Google Scholar

23. Fawzy, GA, Al-Taweel, AM, Perveen, S. Anticancer activity of flavane gallates isolated from Plicosepalus curviflorus. Phcog Mag 2014;10:S519–23, https://doi.org/10.4103/0973-1296.139787.Search in Google Scholar PubMed PubMed Central

24. Fujii, W, Toda, K, Matsumoto, K, Kawaguchi, K, Kawahara, SI, Hattori, Y, et al.. Syntheses of prodelphinidin B1, B2, and B4 and their antitumor activities against human PC-3 prostate cancer cell lines. Tetrahedron Lett 2013;54:7188–92, https://doi.org/10.1016/j.tetlet.2013.10.113.Search in Google Scholar

25. Kuo, PL, Hsu, YL, Lin, TC, Lin, CC. Prodelphinidin B-2 3,3′-di-O-gallate from Myrica rubra inhibits proliferation of A549 carcinoma cells via blocking cell cycle progression and inducing apoptosis. Eur J Pharmacol 2004;501:41–8, https://doi.org/10.1016/j.ejphar.2004.08.024.Search in Google Scholar PubMed

26. Gu, Y, Turck, CW, Morgan, DO. Inhibition of CDK2 activity in vivo by an associated 20K regulatory subunit. Nature 1993;366:707–10, https://doi.org/10.1038/366707a0.Search in Google Scholar PubMed

27. Wade Harper, J, Adami, GR, Wei, N, Keyomarsi, K, Elledge, SJ. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell 1993;75:805–16, https://doi.org/10.1016/0092-8674(93)90499-g.Search in Google Scholar PubMed

28. Nagata, S, Golstein, P. The Fas death factor. Science 1995;267:1449–56, https://doi.org/10.1126/science.7533326.Search in Google Scholar PubMed

29. Zhang, W, Shen, YH, Lou, ZY, Liu, RH, Zhang, C, Fu, P, et al.. Two new flavanes and bioactive compounds from Daphne tangutica Maxim. Nat Prod Res 2007;21:1021–6, https://doi.org/10.1080/14786410701371603.Search in Google Scholar PubMed

30. Ji, S, Tang, S, Li, K, Li, Z, Liang, W, Qiao, X, et al.. Licoricidin inhibits the growth of SW480 human colorectal adenocarcinoma cells in vitro and in vivo by inducing cycle arrest, apoptosis and autophagy. Toxicol Appl Pharmacol 2017;326:25–33, https://doi.org/10.1016/j.taap.2017.04.015.Search in Google Scholar PubMed

31. Ahmed, D, Eide, PW, Eilertsen, IA, Danielsen, SA, Eknæs, M, Hektoen, M, et al.. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2013;2:e71, https://doi.org/10.1038/oncsis.2013.35.Search in Google Scholar PubMed PubMed Central

32. Park, SY, Lim, SS, Kim, JK, Kang, IJ, Kim, JS, Lee, C, et al.. Hexane-ethanol extract of Glycyrrhiza uralensis containing licoricidin inhibits the metastatic capacity of DU145 human prostate cancer cells. Br J Nutr 2010;104:1272–82, https://doi.org/10.1017/s0007114510002114.Search in Google Scholar

33. Park, SY, Kwon, SJ, Lim, SS, Kim, JK, Lee, KW, Park, JHY. Licoricidin, an active compound in the hexane/ethanol extract of Glycyrrhiza uralensis, inhibits lung metastasis of 4T1 murine mammary carcinoma cells. Int J Mol Sci 2016;17:934, https://doi.org/10.3390/ijms17060934.Search in Google Scholar PubMed PubMed Central

34. Ji, S, Liang, WF, Li, ZW, Feng, J, Wang, Q, Qiao, X, et al.. Efficient and selective glucosylation of prenylated phenolic compounds by Mucor hiemalis. RSC Adv 2016;6:20791–9, https://doi.org/10.1039/c6ra00072j.Search in Google Scholar

35. Muthyala, RS, Ju, YH, Sheng, S, Williams, LD, Doerge, DR, Katzenellenbogen, BS, et al.. Equol, a natural estrogenic metabolite from soy isoflavones: convenient preparation and resolution of R- and S-equols and their differing binding and biological activity through estrogen receptors alpha and beta. Bioorg Med Chem 2004;12:1559–67, https://doi.org/10.1016/j.bmc.2003.11.035.Search in Google Scholar PubMed

36. Lu, Z, Zhou, R, Kong, Y, Wang, J, Xia, W, Guo, J, et al.. S-equol, a secondary metabolite of natural anticancer isoflavone daidzein, inhibits prostate cancer growth in vitro and in vivo, though activating the akt/FOXO3a pathway. Curr Cancer Drug Targets 2016;16:455–65, https://doi.org/10.2174/1568009616666151207105720.Search in Google Scholar PubMed

37. Yang, ZP, Zhao, Y, Huang, F, Chen, J, Yao, YH, Li, J, et al.. Equol inhibits proliferation of human gastric carcinoma cells via modulating Akt pathway. World J Gastroenterol 2015;21:10385–99, https://doi.org/10.3748/wjg.v21.i36.10385.Search in Google Scholar PubMed PubMed Central

38. Kim, EY, Shin, JY, Park, YJ, Kim, AK. Equol induces mitochondria-mediated apoptosis of human cervical cancer cells. Anticancer Res 2014;34:4985–92.Search in Google Scholar

39. Hod, R, Maniam, S, Mohd Nor, NH. A systematic review of the effects of equol (soy metabolite) on breast cancer. Molecules 2021;26:1105, https://doi.org/10.3390/molecules26041105.Search in Google Scholar PubMed PubMed Central

40. Zhang, J, Ren, L, Yu, M, Liu, X, Ma, W, Huang, L, et al.. S-equol inhibits proliferation and promotes apoptosis of human breast cancer MCF-7 cells via regulating miR-10a-5p and PI3K/AKT pathway. Arch Biochem Biophys 2019;672:108064, https://doi.org/10.1016/j.abb.2019.108064.Search in Google Scholar PubMed

41. Choi, EJ, Ahn, WS, Bae, SM. Equol induces apoptosis through cytochrome c-mediated caspases cascade in human breast cancer MDA-MB-453 cells. Chem Biol Interact 2009;177:7–11, https://doi.org/10.1016/j.cbi.2008.09.031.Search in Google Scholar PubMed

42. Eun, JC, Kim, T. Equol induced apoptosis via cell cycle arrest in human breast cancer MDA-MB-453 but not MCF-7 cells. Mol Med Rep 2008;1:239–44.Search in Google Scholar

43. Magee, PJ, Allsopp, P, Samaletdin, A, Rowland, IR. Daidzein, R-(+)equol and S-(-)equol inhibit the invasion of MDA-MB-231 breast cancer cells potentially via the down-regulation of matrix metalloproteinase-2. Eur J Nutr 2014;53:345–50, https://doi.org/10.1007/s00394-013-0520-z.Search in Google Scholar PubMed

44. Taghizadeh, B, Ghavami, L, Nikoofar, A, Goliaei, B. Equol as a potent radiosensitizer in estrogen receptor-positive and -negative human breast cancer cell lines. Breast Cancer 2015;22:382–90, https://doi.org/10.1007/s12282-013-0492-0.Search in Google Scholar PubMed

45. Bellou, S, Karali, E, Bagli, E, Al-Maharik, N, Morbidelli, L, Ziche, M, et al.. The isoflavone metabolite 6-methoxyequol inhibits angiogenesis and suppresses tumor growth. Mol Cancer 2012;11:1–11, https://doi.org/10.1186/1476-4598-11-35.Search in Google Scholar PubMed PubMed Central

46. Brown, DM, Kelly, GE, Husband, AJ. Flavonoid compounds in maintenance of prostate health and prevention and treatment of cancer. Mol Biotechnol 2005;30:253–70, https://doi.org/10.1385/mb:30:3:253.10.1385/MB:30:3:253Search in Google Scholar PubMed

47. Li, SR, Chen, PY, Chen, LY, Lo, YF, Tsai, IL, Wang, EC. Synthesis of haginin E, equol, daidzein, and formononetin from resorcinol via an isoflavene intermediate. Tetrahedron Lett 2009;50:2121–3, https://doi.org/10.1016/j.tetlet.2009.02.159.Search in Google Scholar

48. Porter, K, Fairlie, WD, Laczka, O, Delebecque, F, Wilkinson, J. Idronoxil as an anticancer agent: activity and mechanisms. Curr Cancer Drug Targets 2020;20:341–54, https://doi.org/10.2174/1568009620666200102122830.Search in Google Scholar PubMed

49. Li, Y, Huang, X, Huang, Z, Feng, J. Phenoxodiol enhances the antitumor activity of gemcitabine in gallbladder cancer through suppressing Akt/mTOR pathway. Cell Biochem Biophys 2014;70:1337–42, https://doi.org/10.1007/s12013-014-0061-y.Search in Google Scholar PubMed

50. Sapi, E, Alvero, AB, Chen, W, O’Malley, D, Hao, XY, Dwipoyono, B, et al.. Resistance of ovarian carcinoma cells to docetaxel is XIAP dependent and reversible by phenoxodiol. Oncol Res 2004;14:567–78, https://doi.org/10.3727/0965040042707943.Search in Google Scholar PubMed

51. Yao, C, Wu, S, Li, D, Ding, H, Wang, Z, Yang, Y, et al.. Co-administration phenoxodiol with doxorubicin synergistically inhibit the activity of sphingosine kinase-1 (SphK1), a potential oncogene of osteosarcoma, to suppress osteosarcoma cell growth both in vivo and in vitro. Mol Oncol 2012;6:392–404, https://doi.org/10.1016/j.molonc.2012.04.002.Search in Google Scholar PubMed PubMed Central

52. McPherson, RAC, Galettis, PT, De Souza, PL. Enhancement of the activity of phenoxodiol by cisplatin in prostate cancer cells. Br J Cancer 2009;100:649–55, https://doi.org/10.1038/sj.bjc.6604920.Search in Google Scholar PubMed PubMed Central

53. Yaylaci, E, Onen, HI, Saglam, ASY. Phenoxodiol sensitizes metastatic colorectal cancer cells to 5-fluorouracil-and oxaliplatin-induced apoptosis through intrinsic pathway. EXCLI J 2020;19:936–49, https://doi.org/10.17179/excli2020-2042.Search in Google Scholar PubMed PubMed Central

54. Aguero, MF, Facchinetti, MM, Sheleg, Z, Senderowicz, AM. Phenoxodiol, a novel isoflavone, induces G1 arrest by specific loss in cyclin-dependent kinase 2 activity by p53-independent induction of p21WAF1/CIP1. Cancer Res 2005;65:3364–73, https://doi.org/10.1158/0008-5472.can-04-2429.Search in Google Scholar

55. Isono, M, Sato, A, Asano, T, Okubo, K, Asano, T. Evaluation of therapeutic potential of phenoxodiol, a novel isoflavone analog, in renal cancer cells. Anticancer Res 2018;38:5709–16, https://doi.org/10.21873/anticanres.12908.Search in Google Scholar PubMed

56. Kamsteeg, M, Rutherford, T, Sapi, E, Hanczaruk, B, Shahabi, S, Flick, M, et al.. Phenoxodiol – an isoflavone analog – induces apoptosis in chemoresistant ovarian cancer cells. Oncogene 2003;22:2611–20, https://doi.org/10.1038/sj.onc.1206422.Search in Google Scholar PubMed

57. Aguero, MF, Venero, M, Brown, DM, Smulson, ME, Espinoza, LA. Phenoxodiol inhibits growth of metastatic prostate cancer cells. Prostate 2010;70:1211–21, https://doi.org/10.1002/pros.21156.Search in Google Scholar PubMed

58. Yu, F, Watts, RN, Zhang, XD, Borrow, JM, Hersey, P. Involvement of BH3-only proapoptotic proteins in mitochondrial-dependent Phenoxodiol-induced apoptosis of human melanoma cells. Anti Cancer Drugs 2006;17:1151–61, https://doi.org/10.1097/01.cad.0000231484.17063.9a.Search in Google Scholar PubMed

59. Yee, EMH, Cirillo, G, Brandl, MB, Black, DSC, Vittorio, O, Kumar, N. Synthesis of dextran–phenoxodiol and evaluation of its physical stability and biological activity. Front Bioeng Biotechnol 2019;7:1–9, https://doi.org/10.3389/fbioe.2019.00183.Search in Google Scholar PubMed PubMed Central

60. Herst, PM, Davis, JE, Neeson, P, Berridge, MV, Ritchie, DS. The anti-cancer drug, phenoxodiol, kills primary myeloid and lymphoid leukemic blasts and rapidly proliferating T cells. Haematologica 2009;94:928–34, https://doi.org/10.3324/haematol.2008.003996.Search in Google Scholar PubMed PubMed Central

61. Gara, R, Sundram, V, Chauhan, S, Jaggi, M. Anti-cancer potential of a novel SERM ormeloxifene. Curr Med Chem 2013;20:4177–84, https://doi.org/10.2174/09298673113209990197.Search in Google Scholar PubMed PubMed Central

62. Pillai, LS, Regidi, S, Varghese, SD, Ravindran, S, Maya, V, Varghese, J, et al.. Nonhormonal selective estrogen receptor modulator 1-(2-[4-{(3R,4S)-7-Methoxy-2, 2-dimethyl-3-phenyl-chroman-4yl}phenoxy]ethyl)pyrrolidine hydrochloride (ormeloxifene hydrochloride) for the treatment of breast cancer. Drug Dev Res 2018;79:275–86, https://doi.org/10.1002/ddr.21440.Search in Google Scholar PubMed

63. Khan, S, Shukla, S, Sinha, S, Lakra, AD, Bora, HK, Meeran, SM. Centchroman suppresses breast cancer metastasis by reversing epithelial-mesenchymal transition via downregulation of HER2/ERK1/2/MMP-9 signaling. Int J Biochem Cell Biol 2015;58:1–16, https://doi.org/10.1016/j.biocel.2014.10.028.Search in Google Scholar PubMed

64. Hafeez, BB, Ganju, A, Sikander, M, Kashyap, VK, Hafeez, ZB, Chauhan, N, et al.. Ormeloxifene suppresses prostate tumor growth and metastatic phenotypes via inhibition of oncogenic β-catenin signaling and EMT progression. Mol Cancer Therapeut 2017;16:2267–80, https://doi.org/10.1158/1535-7163.mct-17-0157.Search in Google Scholar PubMed PubMed Central

65. Srivastava, VK, Gara, RK, Bhatt, MLB, Sahu, DP, Mishra, DP. Centchroman inhibits proliferation of head and neck cancer cells through the modulation of PI3K/mTOR Pathway. Biochem Biophys Res Commun 2011;404:40–5, https://doi.org/10.1016/j.bbrc.2010.11.049.Search in Google Scholar PubMed

66. Khan, S, Shukla, S, Farhan, M, Sinha, S, Lakra, AD, Penta, D, et al.. Centchroman prevents metastatic colonization of breast cancer cells and disrupts angiogenesis via inhibition of RAC1/PAK1/β-catenin signaling axis. Life Sci 2020;256:117976, https://doi.org/10.1016/j.lfs.2020.117976.Search in Google Scholar PubMed

67. Pushpakom, S, Iorio, F, Eyers, PA, Escott, KJ, Hopper, S, Wells, A, et al.. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov 2018;18:41–58, https://doi.org/10.1038/nrd.2018.168.Search in Google Scholar PubMed

68. Kemnitzer, W, Drewe, J, Jiang, S, Zhang, H, Wang, Y, Zhao, J, et al.. Discovery of 4-aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay. 1. Structure-activity relationships of the 4-aryl group. J Med Chem 2004;47:6299–310, https://doi.org/10.1021/jm049640t.Search in Google Scholar PubMed

69. Kemnitzer, W, Kasibhatla, S, Jiang, S, Zhang, H, Zhao, J, Jia, S, et al.. Discovery of 4-aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay. 2. Structure-activity relationships of the 7- and 5-, 6-, 8-positions. Bioorg Med Chem Lett 2005;15:4745–51, https://doi.org/10.1016/j.bmcl.2005.07.066.Search in Google Scholar PubMed

70. Kemnitzer, W, Drewe, J, Jiang, S, Zhang, H, Zhao, J, Crogan-Grundy, C, et al.. Discovery of 4-aryl-4 H -chromenes as a new series of apoptosis inducers using a cell- and caspase-based high-throughput screening assay. 3. Structure−Activity relationships of fused rings at the 7,8-positions. J Med Chem 2007;50:2858–64, https://doi.org/10.1021/jm070216c.Search in Google Scholar PubMed

71. Kemnitzer, W, Drewe, J, Jiang, S, Zhang, H, Crogan-Grundy, C, Labreque, D, et al.. Discovery of 4-aryl-4 H -chromenes as a new series of apoptosis inducers using a cell- and caspase-based high throughput screening assay. 4. Structure–Activity relationships of N -alkyl substituted pyrrole fused at the 7,8-positions. J Med Chem 2008;51:417–23, https://doi.org/10.1021/jm7010657.Search in Google Scholar PubMed

72. Kasibhatla, S, Gourdeau, H, Meerovitch, K, Drewe, J, Reddy, S, Qiu, L, et al.. Discovery and mechanism of action of a novel series of apoptosis inducers with potential vascular targeting activity. Mol Cancer Therapeut 2004;3:1365–74.10.1158/1535-7163.1365.3.11Search in Google Scholar

73. Gourdeau, H, Leblond, L, Hamelin, B, Desputeau, C, Dong, K, Kianicka, I, et al.. Antivascular and antitumor evaluation of 2-amino-4-(3-bromo-4,5-dimethoxy-phenyl)-3-cyano-4H-chromenes, a novel series of anticancer agents. Mol Cancer Therapeut 2004;3:1375–83.10.1158/1535-7163.1375.3.11Search in Google Scholar

74. Hinnen, P, Eskens, FALM. Vascular disrupting agents in clinical development. Br J Cancer 2007;96:1159–65, https://doi.org/10.1038/sj.bjc.6603694.Search in Google Scholar PubMed PubMed Central

75. Smolarczyk, R, Czapla, J, Jarosz-Biej, M, Czerwinski, K, Cichoń, T. Vascular disrupting agents in cancer therapy. Eur J Pharmacol 2021;891:173692, https://doi.org/10.1016/j.ejphar.2020.173692.Search in Google Scholar PubMed

76. Smith, CW, Bailey, JM, Billingham, MEJ, Chandrasekhar, S, Dell, CP, Harvey, AK, et al.. The anti-rheumatic potential of a series of 2,4-di-substituted-4H-naphtho[1,2-b]pyran-3-carbonitriles. Bioorg Med Chem Lett 1995;5:2783–8, https://doi.org/10.1016/0960-894x(95)00487-e.Search in Google Scholar

77. Egeblad, M, Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer 2002;2:161–74, https://doi.org/10.1038/nrc745.Search in Google Scholar PubMed

78. Kemnitzer, W, Jiang, S, Wang, Y, Kasibhatla, S, Crogan-Grundy, C, Bubenik, M, et al.. Discovery of 4-aryl-4H-chromenes as a new series of apoptosis inducers using a cell- and caspase-based HTS assay. Part 5: modifications of the 2- and 3-positions. Bioorg Med Chem Lett 2008;18:603–7, https://doi.org/10.1016/j.bmcl.2007.11.078.Search in Google Scholar PubMed

79. Shestopalov, AM, Litvinov, YM, Rodinovskaya, LA, Malyshev, OR, Semenova, MN, Semenov, VV. Polyalkoxy substituted 4H-chromenes: synthesis by domino reaction and anticancer activity. ACS Comb Sci 2012;14:484–90, https://doi.org/10.1021/co300062e.Search in Google Scholar PubMed

80. Chernysheva, NB, Tsyganov, DV, Philchenkov, AA, Zavelevich, MP, Kiselyov, AS, Semenov, RV, et al.. Synthesis and comparative evaluation of 4-oxa- and 4-aza-podophyllotoxins as antiproliferative microtubule destabilizing agents. Bioorg Med Chem Lett 2012;22:2590–3, https://doi.org/10.1016/j.bmcl.2012.01.128.Search in Google Scholar PubMed

Published Online: 2022-02-02

© 2022 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 24.5.2024 from https://www.degruyter.com/document/doi/10.1515/psr-2021-0033/html
Scroll to top button