Letter to the editor

Recent developments on potential new applications of emetine as anti-cancer agent

Philip F. Uzor1[*]

1Department of Pharmaceutical and Medicinal Chemistry, University of Nigeria, Nsukka, 410001, Nigeria

EXCLI J 2016;15:Doc323

 

Dear Editor,

Cancer remains one of the leading causes of global morbidity and mortality, with approximately 14 million new cases and 8.2 million cancer related deaths in 2012 (Stewart and Wild, 2014[37]). Treatment protocols include radiation, surgery, chemotherapy, hormone therapy, immunotherapy and targeted therapy (American Cancer Society, 2015[6]). While chemotherapy is one of the key strategies against cancer, the available drugs are frequently fraught with toxicity and increased frequency of tumor relapse (Gaziano et al., 2016[12]). This calls for an urgent need for more effective anti-tumor agents especially from phytochemicals which are known to be of lower toxicity and cost (Reddy et al., 2003[35]). A wide variety of phytochemicals, particularly alkaloids, have been investigated in recent times in the quest for more effective and safer antitumor agents (Lu et al., 2012[26]; Kharwar et al., 2011[20]). Interestingly, several important anti-tumor alkaloidal drugs have been isolated from medicinal plants including the vinca alkaloids, vinblastine and vincristine, isolated from the Madagascar periwinkle, Catharanthus roseus (Noble et al., 1958[31]; Johnson et al., 1959[17]; Svoboda, 1961[40]) as well as paclitaxel, isolated from Taxus brevifolia (Wani et al., 1971[44]). One effective strategy employed by scientists in this regard is the investigation of known drugs for novel biological effects, the so called 'drug repositioning'. One of such known drugs that have been shown to possess anti-tumor activity is the alkaloidal amoebicidal drug, emetine (EMT).

EMT, chemically designated as 2S,3R,11bS)-2-{[(1R)-6,7-dimethoxy-1,2,3,4-tetrahydroisoquinolin-1-yl]methyl}-3-ethyl-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido [2,1-a]isoquinoline (Figure 1(Fig. 1)), is an isoquinoline alkaloid which occurs in the families of Alangiaceae, Icacinaceae, and Rubiaceae. The major source of EMT and its analogs is Psychotria ipecacuanha Stokes (Rubiaceae) which is also known as Cephaelis ipecacuanha A. Rich (ipecac) where it is the principal alkaloid (Wiegrebe et al., 1984[45]). It is clinically used (as a dihydrochloride) in the treatment of amoebiasis, a protozoan infection (Vedder, 1912[42]) and it has emetic properties. It is reportedly a protein synthesis inhibitor in eukaryotes (Grollman, 1968[13]). The biosynthesis of EMT and cephaeline (another alkaloid found in ipecac) comes from two main biosynthesis pathways, the biosynthesis of dopamine from L-tyrosine and that of secologanin from geranyl diphosphate (Cheong et al., 2011[9]; Nomura et al., 2010[32]).

The anti-cancer effect of EMT was first reported on malignant human tumors in 1918 by Lewisohn (1918[25]) but since he was unable to reproduce this effect in laboratory animals, he concluded that the drug had no anti-tumor properties and that the tumor regression must have been spontaneous. However, in the following year, Van Hoosen (1919[41]) further reported the remission of various malignancies in a number of patients by EMT. This is followed in later years by reports of effectiveness of EMT in rat Yoshida sarcoma (Isaka, 1950[16]), intra-abdominal and retroperitoneal nonspecific granulomas (Grollman, 1965[13]) and in murine leukemia (Jondorf et al., 1970[18]). Besides, the potency of an analogue of EMT, dehydroemetine, was also shown in chronic granulocytic leukemia (Abd-Rabbo, 1966[2]), various malignancies (Abd-Rabbo, 1969[1]) as well as in Hodgkin's disease and rectal adenocarcinoma (Wyburn-Mason, 1966[46]). Based on these reports, phase I and II clinical trials with EMT were done in the early 1970s (Panettiere and Coltman, 1971[34]; Street, 1972[38]; Mastrangelo et al., 1973[27]; Siddiqui et al., 1973[36]; Moertel et al., 1974[29]; Kane et al., 1975[19]). The drug was, however, discontinued from the clinical trials (Von Hoff et al., 1977[43]) due to its very narrow therapeutic index, cardiac toxicity and other adverse effects which were also observed in the treatment of amoebic patients (Knight, 1980[22]). Since then the drug has been used in in vitro experimental studies requiring inhibition of protein biosynthesis (Akinboye et al., 2012[5]). The data from these recent studies have further shown EMT as a modulator of different cancer related biological pathways. In fact, excellent review by Akinboye and Bakare (2011[3]) has shown that EMT exhibits its anti-tumor effect by apoptosis through such mechanisms as inhibition of protein biosynthesis, DNA interaction and regulation of pro-apoptotic factors. In more recent years also, various studies have further investigated the role of EMT in cancer growth arrest and its biological targets using a variety of human carcinoma cell lines. New derivatives have also been synthesized and reported to be efficacious but less toxic to normal cells. Also the drug has been investigated in combination with other agents to assess their anti-tumor synergistic effect which will warrant reduction in its dose. These studies are geared towards bringing back EMT or its derivatives to the clinical limelight in cancer chemotherapy. The present report summarizes these more recent anti-tumor updates on EMT (Table 1(Tab. 1); References in Table 1: Sun et al., 2015[39]; Kim et al., 2015[21]; Mayank and Jaitak, 2016[28]; Han et al., 2014[15]; Myhren et al., 2014[30]; Foreman et al., 2013[10]; Akinboye et al., 2012[5]; Larrson et al., 2012[24]; Pan et al., 2011[33]; Kong et al., 2010[23]). It is hoped that this report will further spur research interests on EMT and its structural modifications towards potential application in cancer chemotherapy.

 

References

1. Abd-Rabbo H. Chemotherapy of neoplasia (cancer) with dehydroemetine. J Trop Med Hyg. 1969;72:287-90.
2. Abd-Rabbo H. Dehydroemetine in chronic leukemia. Lancet. 1966;1:1161-2.
3. Akinboye ES, Bakare O. Biological activities of emetine. Open Nat Prod J. 2011;4:8-15.
4. Akinboye ES, Brennen WN, Rosen DM, Bakare O, Denmeade SR. Iterative design of emetine-based prodrug targeting fibroblast activation protein (FAP) and dipeptidyl peptidase IV DPPIV using a tandem enzymatic activation strategy. Prostate. 2016;76:703-14.
5. Akinboye ES, Rosen MD, Denmeade SR, Kwabi-Addo B, Bakare O. Design, synthesis and evaluation of pH-dependent hydrolysable emetine analogs as treatment for prostate cancer. J Med Chem. 2012;55:7450–9.
6. American Cancer Society. Cancer facts & figures 2015. Atlanta: American Cancer Society, 2015.
7. Bamji ZD, Washington KN, Akinboye E, Bakare O, Kanaan YM, Copeland RL Jr. Apoptotic effects of novel dithiocarbamate analogs of emetine in prostate cancer cell lines. Anticancer Res. 2015;35:4723-32.
8. Bamji, ZD. Dithiocarbamate analogs of emetine as potential chemotherapeutic agents: their apoptotic role, gene regulation and efficacy in prostate cancer. PhD Thesis. Washington, DC: Howard University, 2011.
9. Cheong BE;Takemura T;Yoshimatsu K, Sato F. Molecular cloning of an O-methyltransferase from adventitious roots of Carapichea ipecacuanha. Biosci Biotechnol Biochem. 2011;75:107–13.
10. Foreman K, Jesse J, Gupta G, Maywood, IL. Emetine dihydrochloride: a novel therapy for bladder urothelial carcinoma. J Urol. 2013;189:e245.
11. Foreman KE, Patel D, Davidson V, Kuo P, Flanigan R, Gupta GN, et al. MP45-09 emetine dihydrochloride preferentially inhibits Hif1α and Hif2α expression in bladder cancer cells. J Urol. 2015;193:e538–9.
12. Gaziano R, Moroni G, Buè C, Miele MT, Sinibaldi-Vallebona P, Pica F. Antitumor effects of the benzophenanthridine alkaloid sanguinarine: Evidence and perspectives. World J Gastrointest Oncol. 2016;8:30-9.
13. Grollman AI. Emetine in the treatment of intra-abdominal and retroperitoneal nonspecific granulomas. Surg Gynec Obstet. 1965;120:792-6.
14. Grollman AP. Inhibitors of protein biosynthesis. V. Effects of emetine on protein and nucleic acid biosynthesis in HeLa cells. J Biol Chem. 1968;243:4089-94.
15. Han Y, Park S, Kinyua AW, Andera L, Kim KW, Kim I. Emetine enhances the tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis of pancreatic cancer cells by downregulation of myeloid cell leukemia sequence-1 protein. Oncol Rep. 2014;31:456-62.
16. Isaka H. The effect of emetine hydrochloride upon the Yoshida sarcoma. GANN. 1950;41:165-8.
17. Johnson IS, Wright HF, Svoboda GH. Experimental basis for clinical evaluation of anti-tumor principles derived from Vinca rosea Linn. J Lab Clin Med. 1959;54:830.
18. Jondorf WR, Abbott BJ, Greenberg NA, Mead JAR. Antileukemic effectiveness of (-)-emetine and some of its derivatives. Pharmacologist. 1970;12:282.
19. Kane RC, Cohen MH, Broder LE, Bull MI, Creaven PJ, Fossieck BE Jr. Phase I-II evaluation ofemetine (NSC-33 669) in the treatment of epidermoid bronchogenic carcinoma. Cancer Chemother Rep. 1975;59:1171–2.
20. Kharwar RN, Mishra A, Gond SK, Stierle A, Stierle D. Anticancer compounds derived from fungal endophytes: their importance and future challenges. Nat Prod Rep. 2011;28:1208-28.
21. Kim JH, Cho EB, Lee J, Jung O, Ryuc BJ, Kim SH, et al. Emetine inhibits migration and invasion of human non-small-cell lung cancer cells via regulation of ERK and p38 signaling pathways. Chem Biol Interact. 2015;242:25–33.
22. Knight R. The chemotherapy of amoebiasis. J Antimicrob Chemother. 1980;6:577–93.
23. Kong HS, Lee S, Beebe K, Scroggins B, Gupta G, Lee MJ, et al. Emetine promotes von Hippel-Lindau-independent degradation of hypoxia-inducible factor-2 in clear cell renal carcinoma. Mol Pharmacol. 2010;78:1072-8.
24. Larsson DE, Hassan SB, Oberg K, Granberg D. The cytotoxic effect of emetine and CGP-74514A studied with the hollow fiber model and ArrayScan assay in neuroendocrine tumors in vitro. Anticancer Agents Med. 2012;12:783-90.
25. Lewisohn R. Action of emetine on malignant tumors. J Am Med Assoc. 1918;70:9-10.
26. Lu J-J, Bao J-L, Chen X-P, Huang M, Wang Y-T. Alkaloids isolated from natural herbs as the anticancer agents. Evidence-based Compl Altern Med. 2012;2012:article ID 485042.
27. Mastrangelo MJ, Grage TB, Bellet RE, Weiss AJ. A phase I study of emetine hydrochloride (NSC-33 669) in solid tumors. Cancer. 1973;31:1170–5.
28. Mayank JV. Molecular docking study of natural alkaloids as multi-targeted hedgehog pathway inhibitors in cancer stem cell therapy. Computat Biol Chem. 2016;62:145-54.
29. Moertel CG, Schutt AJ, Hahn RG, Reitemeier RJ. Treatment of advanced gastrointestinal cancer with emetine (NSC-33 669). Cancer Chemother Rep. 1974;58:229–32.
30. Myhren L, Nilssen IM, Nicolas V, Døskeland SO, Barratt G, Herfindal L. Efficacy of multi-functional liposomes containing daunorubicin and emetine for treatment of acute myeloid leukaemia. Eur J Pharm Biopharmaceut. 2014;88:186-93.
31. Noble RL, Beer CT, Cutts JH. Role of chance observations in chemotherapy: Vinca rosea. Ann NY Acad Sci. 1958;76:882-94.
32. Nomura T, Kutchan TM. Three new O-methyltransferases are sufficient for all o-methylation reactions of ipecac alkaloid biosynthesis in root culture of psychotria ipecacuanha. J Biol Chem. 2010;285:7722–38.
33. Pan D, Boon-Unge K, Govitrapong P, Zhou J. Emetine regulates the alternative splicing of caspase 9 in tumor cells. Oncol Lett. 2011;2:1309-12.
34. Panettiere F, Coltman CA Jr. Phase 1 experience with emetine hydrochloride (NSC-33 669) as an antitumor agent. Cancer. 1971;27:835–41.
35. Reddy L, Odhav B, Bhoola KD. Natural products for cancer prevention: a global perspective. Pharmacol Ther. 2003;99:1-13.
36. Siddiqui S, Firat D, Olshin S. Phase II study of emetine (NSC-33 669) in the treatment of solid tumors. Cancer Chemother Rep. 1973;57:423–8.
37. Stewart BW, Wild CP. World Cancer Report 2014. Geneva: IARC, 2014.
38. Street EW. Cyclophosphamide plus emetine in lung cancer. Lancet. 1972;2:281–2.
39. Sun Q, Yogosawa S, Iizumi Y, Sakai T, Sowa Y. The alkaloid emetine sensitizes ovarian carcinoma cells to cisplatin through downregulation of bcl-xL. Int J Oncol. 2015;46:389-94.
40. Svoboda GH. Alkaloids of Vinca rosea (Catharanthus roseus). IX. Extraction and characterization of leurosidine and leurocristine. Lloydia. 1961;24:173-8.Van Hoose B. Emetine hydrochloride in malignancy. Women’s Med J. 1919;29:102-16.
41. Van Hoose B. Emetine hydrochloride in malignancy. Women's Med J. 1919;29:102-16.
42. Vedder EB. An experimental study of the action of Ipecacuanha on Amebae. J Trop Med Hyg. 1912;15:313.
43. Von Hoff DD, Rozencweig M, Soper WT, Helman LJ, Penta JS, Davis HL, et al. Whatever happened to NSC--? An analysis of clinical results of discontinued anticancer agents. Cancer Treat Rep. 1977;61:759-68.
44. Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. Plant antitumor agents. VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J Am Chem Soc. 1971;93:2325–7.
45. Wiegrebe W, Kramer WJ, Shamma M. The emetine alkaloids. J Nat Prod. 1984;47:397-408.
46. Wyburn-Mason R. Dehydroemetine in chronic leukaemia. Lancet. 1966;1:1266-7.
 
 

Figure 1: Chemical structure of EMT

 

Table 1: Recent studies on EMT in relation to anti-cancer effect

[*] Corresponding Author:

Philip F. Uzor, Department of Pharmaceutical and Medicinal Chemistry, University of Nigeria, Nsukka, 410001, Nigeria; Tel: +234 8037008294, eMail: philip.uzor@unn.edu.ng