6
4-Aminoquinoline-Triazine-Based Hybrids with Improved In Vitro Antimalarial Activity Against CQ-Sensitive and CQ-Resistant Strains of Plasmodium falciparum Sunny Manohar 1 , Shabana I. Khan 2 and Diwan S. Rawat 1, * 1 Department of Chemistry, University of Delhi, Delhi, 110007, India 2 National Centre for Natural Products Research, University of Mississippi, Oxford, MS 38677, USA *Corresponding author: Diwan S. Rawat, [email protected] A systematic chemical modification in the triazine moiety covalently attached via suitable linkers to 4-amino-7-chloroquinolines yielded a series of new 7-chloro-4-aminoquinoline-triazine hybrids exhibiting high in vitro activity against W2 (chloroquine-resistant) and D6 (chloroquine-sensitive) strains of Plasmodium falciparum without any toxicity against mammalian cell lines (Vero, LLC-PK11, HepG2). Many of the com- pounds (6, 8, 10, 11, 13, 14, 16, 27, 29 and 33) showed excellent potency against chloroquine sensitive and resistant strains. In particular, compounds 6, 8, 14, 16 and 29 were found to be significantly more active than chloroquine against the chloroquine-resistant strains (W2 clone) of P. falciparum. Key words: antimalarial, hybrids, Plasmodium falciparum Received 11 August 2012, revised 18 December 2012 and accepted for publication 8 January 2013 The prevalence of malaria has been documented about 5000 years back, but still it has remained one of the most lethal disease causing deaths of over one million people every year (1,2). The drugs once considered to be safe and effective have become useless due to the incidences of increasing reports of drug resistance and that has put an enormous burden on public health (3,4). Among the most commonly used drugs, chloroquine (CQ) (3) had been a drug of choice for the treatment of malaria for nearly half a century and considered to be a wonder drug due to the fact that it was effective, easily available, safe and inexpensive (5). The added advantage was that it could be administered to infants and pregnant women. The problem has been complicated due to the fact that P. falciparum, the cause of the most lethal variety of malaria, has developed resistance against CQ and other antimalarials. Number of isolates of P. falciparum have developed resistance against not only CQ but against the majority of currently used antimalarials. To address the issue of drug resistance, several approaches that include chloroquine-resistant (CQR) reversal agents (6,7), com- bination therapy (8) and use of endoperoxide-based antimalarials have been introduced (914). Although end- operoxides are considered to be useful against resistant strains, but these compounds are less affordable, and recently, resistance against endoperoxide-derived anti- malarials has already been reported (15). Arguably, amino- quinolines such as quinine, CQ, mefloquine and amodiaquine are among the most successful antimalarial drugs ever used (5,16,17), and structural modification of these molecules has resulted many additional lead compounds with improved activity against CQR strains (18 20). Structure activity relationship studies revealed that 4-aminoquinolines carrying an aliphatic side chain with 23 or 1012 carbon, and the incorporation of a phenol moiety is well tolerated and afford excellent activity-toxicity profiles (2024). Many reports revealed that replacement of ethyl group by isopropyl or tert-butyl groups can furnish meta- bolically more stable antimalarials with enhanced lifetime and retained activity against drug-resistant strains of P. fal- ciparum (25,26). To develop safe, effective and inexpen- sive antimalarials that are active against resistant strains and multiple species of Plasmodia, efforts have therefore been directed towards the development of new CQ ana- logues. Towards these goals, concept of hybrid molecules have been put forward by Meunier and co-workers (27) in malaria chemotherapy, in which two antimalarial pharma- cophore are attached covalently, and it is anticipated that these kinds of molecules may solve the problem of drug resistance. In the past few years, number of 4-aminoquin- oline-based hybrid molecules are synthesised in an attempt to solve the problem of drug resistance (2736). The previous studies from our laboratory (32,33) and oth- ers showed that aminoquinoline-triazine hybrids exhibit potent antimalarial activity (37,38). In continuation to our efforts towards the development of new antimalarials (3942), we report herein synthesis and antimalarial activity of ª 2013 John Wiley & Sons A/S. doi: 10.1111/cbdd.12108 625 Chem Biol Drug Des 2013; 81: 625–630 Research Article

4-Aminoquinoline-Triazine-Based Hybrids with Improved In Vitro Antimalarial Activity Against CQ-Sensitive and CQ-Resistant Strains of Plasmodium falciparum

  • Upload
    du-in

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

4-Aminoquinoline-Triazine-Based Hybrids withImproved In Vitro Antimalarial Activity AgainstCQ-Sensitive and CQ-Resistant Strains ofPlasmodium falciparum

Sunny Manohar1, Shabana I. Khan2 andDiwan S. Rawat1,*

1Department of Chemistry, University of Delhi, Delhi,110007, India2National Centre for Natural Products Research, Universityof Mississippi, Oxford, MS 38677, USA*Corresponding author: Diwan S. Rawat,[email protected]

A systematic chemical modification in the triazinemoiety covalently attached via suitable linkers to4-amino-7-chloroquinolines yielded a series of new7-chloro-4-aminoquinoline-triazine hybrids exhibitinghigh in vitro activity against W2 (chloroquine-resistant)and D6 (chloroquine-sensitive) strains of Plasmodiumfalciparum without any toxicity against mammalian celllines (Vero, LLC-PK11, HepG2). Many of the com-pounds (6, 8, 10, 11, 13, 14, 16, 27, 29 and 33) showedexcellent potency against chloroquine sensitive andresistant strains. In particular, compounds 6, 8, 14, 16and 29 were found to be significantly more active thanchloroquine against the chloroquine-resistant strains(W2 clone) of P. falciparum.

Key words: antimalarial, hybrids, Plasmodium falciparum

Received 11 August 2012, revised 18 December 2012 andaccepted for publication 8 January 2013

The prevalence of malaria has been documented about5000 years back, but still it has remained one of the mostlethal disease causing deaths of over one million peopleevery year (1,2). The drugs once considered to be safeand effective have become useless due to the incidencesof increasing reports of drug resistance and that has putan enormous burden on public health (3,4). Among themost commonly used drugs, chloroquine (CQ) (3) hadbeen a drug of choice for the treatment of malaria fornearly half a century and considered to be a wonder drugdue to the fact that it was effective, easily available, safeand inexpensive (5). The added advantage was that itcould be administered to infants and pregnant women.The problem has been complicated due to the fact that

P. falciparum, the cause of the most lethal variety ofmalaria, has developed resistance against CQ and otherantimalarials. Number of isolates of P. falciparum havedeveloped resistance against not only CQ but against themajority of currently used antimalarials. To address theissue of drug resistance, several approaches that includechloroquine-resistant (CQR) reversal agents (6,7), com-bination therapy (8) and use of endoperoxide-basedantimalarials have been introduced (9–14). Although end-operoxides are considered to be useful against resistantstrains, but these compounds are less affordable, andrecently, resistance against endoperoxide-derived anti-malarials has already been reported (15). Arguably, amino-quinolines such as quinine, CQ, mefloquine andamodiaquine are among the most successful antimalarialdrugs ever used (5,16,17), and structural modification ofthese molecules has resulted many additional leadcompounds with improved activity against CQR strains (18–20). Structure activity relationship studies revealed that4-aminoquinolines carrying an aliphatic side chain with 2–3or 10–12 carbon, and the incorporation of a phenol moietyis well tolerated and afford excellent activity-toxicity profiles(20–24). Many reports revealed that replacement of ethylgroup by isopropyl or tert-butyl groups can furnish meta-bolically more stable antimalarials with enhanced lifetimeand retained activity against drug-resistant strains of P. fal-ciparum (25,26). To develop safe, effective and inexpen-sive antimalarials that are active against resistant strainsand multiple species of Plasmodia, efforts have thereforebeen directed towards the development of new CQ ana-logues. Towards these goals, concept of hybrid moleculeshave been put forward by Meunier and co-workers (27) inmalaria chemotherapy, in which two antimalarial pharma-cophore are attached covalently, and it is anticipated thatthese kinds of molecules may solve the problem of drugresistance. In the past few years, number of 4-aminoquin-oline-based hybrid molecules are synthesised in anattempt to solve the problem of drug resistance (27–36).

The previous studies from our laboratory (32,33) and oth-ers showed that aminoquinoline-triazine hybrids exhibitpotent antimalarial activity (37,38). In continuation to ourefforts towards the development of new antimalarials (39–42), we report herein synthesis and antimalarial activity of

ª 2013 John Wiley & Sons A/S. doi: 10.1111/cbdd.12108 625

Chem Biol Drug Des 2013; 81: 625–630

Research Article

series of new 4-aminoquinoline-triazine-based hybrids.Systematic chemical modification in the triazine ring andlinker led to the discovery of many potent antimalarialagents.

Results and Discussion

ChemistrySynthesis of substituted 4-aminoquinolines (2a–c) wasachieved by aromatic nucleophillic substitution (SNAr) of4,7-dichloroquinoline (1) with an excess of different linear-chained aliphatic diamines in neat conditions as reportedin literature (Scheme 1) (43).

Temperature controlled stepwise displacement of chlorineatom from commercially available starting material cyanuricchloride (3) was accomplished in stepwise to achieve thesynthesis of target compounds (6–33) as outlined inScheme 2. In the first step, cyanuric chloride (3) wassubjected to nucleophillic substitution by piperidine ormorpholine in presence of a base at 0 °C to yield

monosbustituted triazines (4a,b). These monosubstitutedtriazines (4a,b) on reaction with various aliphatic and aro-matic amines at room temperature provided disubstitutedtriazines (5a–j) in good yield. Third chlorine of disubstitutedtriazines (5a–j) was then substituted by 4-aminoquinolineshaving varying alkyl chain lengths (2a–c) at elevatedtemperature to yield final trisubstituted triazines (6–33) inmoderate to excellent yields.

Antimalarial activity and cytotoxicity evaluationThe synthesized compounds (6–33) were evaluated fortheir in vitro antimalarial activity against D6 clone (CQ-sen-sitive) and W2 clone (CQ-resistant) of P. falciparum

(Table 1), while the cytotoxicity evaluation was carriedagainst a panel of three mammalian cells (Vero, LLC-PK11

and HepG2) by using doxorubicin as standard drug(Table 2). Selectivity index of antimalarial activity was cal-culated based on the cytotoxicity to Vero cells. Of 28compounds synthesised in the present study, 10 com-pounds (6, 8, 10, 11, 13, 14, 16, 27, 29 and 33) haveshown better antimalarial activity (IC50 ranging from 0.11to 0.42 lM) than CQ, while three compounds (12, 26 and32) have shown comparable activity (IC50 ranging from0.48 to 0.49 lM) to CQ against W2 clone of P. falciparum.Four of these hybrids (17, 26, 29 and 31) have also shownpotent antimalarial activity (IC50 ranging from 0.06 to0.10 lM) with high selective index against D6 clone ofP. falciparum. Structure activity relationship studies ofthese hybrid shows that the spacer which links 4-amino-quinoline to 1,3,5-triazine moiety plays an important role indefining the activity of these molecules. It is evident fromthe Table 1 that increasing the length of the linker from C2to C4, activity of the resulting compounds increases.

Scheme 1: (a) aliphatic diamines, neat, 110–120 °C, 6–8 h, 80–85%.

Scheme 2: (a) morpholine or piperidine, K2CO3, THF, 0–5 °C, 3 h, 75–80% (b) aromatic or aliphatic amines, K2CO3, THF, rt, 3 h, 70–85%; (c) 2a–c, K2CO3, DMF, 100–110 °C, 10–12 h, 65–85%.

626 Chem Biol Drug Des 2013; 81: 625–630

Manohar et al.

However, some deviations from this trend were alsoobserved with few compounds (7, 13, 19, 25 against D6clone and 7, 16, 19, 31 against W2 clone). Enhanced anti-malarial activity with high selective index against W2 cloneof P. falciparum has been observed for compoundscontaining aromatic substitution on 1,3,5-triazine (seven of

12 compounds are better than CQ) than their aliphaticcounterparts (two of 16 compounds are better than CQ).This may be attributed to greater lipophilic character asso-ciated with aromatic compounds (Table 1), which is ingood agreement to our previous observation (33). It maybe noted that no obvious change in activity pattern was

Table 1: In vitro antimalarial activity of new 4-aminoquinoline-triazine conjugates.

NN

N6-33

N N NX

n

R

HHN

CI

Entry n X R cLogP*

P. falciparum (D6 Clone)P. falciparum (W2Clone)

IC50 (lM) S. I. IC50 (lM) S. I.

6 1 O Aniline 6.36 0.29 >86.2 0.17 >147.07 2 O Aniline 6.77 0.44 >56.8 1.05 >23.88 3 O Aniline 6.88 0.29 >170.6 0.25 >198.09 1 O 4-ethyl aniline 7.39 0.47 >53.1 0.67 >37.3

10 2 O 4-ethyl aniline 7.79 0.40 72.0 0.42 68.511 3 O 4-ethyl aniline 7.91 0.39 >64.1 0.18 >138.812 1 O 4-fluoro aniline 6.52 0.24 >104.1 0.48 >52.013 2 O 4-fluoro aniline 6.92 0.43 >58.1 0.33 >75.714 3 O 4-fluoro aniline 7.04 0.24 >104.1 0.11 >227.215 1 O 4-methoxy aniline 6.29 0.25 >100.0 0.59 >42.316 2 O 4-methoxy aniline 6.69 0.16 >156.2 0.15 >166.617 3 O 4-methoxy aniline 6.81 0.07 >357.1 0.71 >35.218 1 O HN

OH3.54 0.49 >51.0 1.70 >14.7

19 2 O HNOH

4.02 0.50 >50.0 2.06 >12.1

20 3 O HNOH

4.14 0.19 >131.5 0.59 >42.3

21 1 O HN OH 3.99 0.67 >37.3 1.56 >16.0

22 2 O HN OH 4.39 0.63 >39.6 1.52 >16.4

23 3 O HN OH 4.42 0.22 >113.6 0.57 >43.8

24 1 O HNOH

3.88 0.48 >52.0 1.52 >16.4

25 2 O HNOH

4.39 0.51 >49.0 1.12 >22.3

26 3 O HNOH

4.50 0.09 >277.7 0.49 >51.0

27 1 CH2 HNOH

5.52 0.14 >178.5 0.40 >62.5

28 2 CH2 HNOH

5.92 ND ND ND ND

29 3 CH2 HNOH

6.04 0.10 >250.0 0.28 >89.2

30 1 CH2 HN OH 5.44 0.21 >119.0 0.83 >30.1

31 3 CH2 HN OH 6.05 0.06 >858.3 1.17 >44.0

32 1 CH2 HNOH

5.16 0.19 >131.5 0.49 >51.0

33 3 CH2 HNOH

5.60 0.19 >131.5 0.42 >59.5

Chloroquine (CQ) 0.05 >500 0.43 >58.1Artemisinin 0.015 >1666.6 0.014 >1785.7

ND – not determined; Selectivity index (SI) – IC50 for cytotoxicity towards VERO cells/IC50 for antimalarial activity; cLogP – Calculated byCHEMBIODRAW 11 Software, Cambridge, MA, USA.

Chem Biol Drug Des 2013; 81: 625–630 627

4-Aminoquinoline-triazine Based Hybrids

observed by changing the substituents (fluoro, ethyl ormethoxy groups) on phenyl ring attached to 1,3,5-triazinemoiety. Most interestingly, the introduction of aminoalcohols with terminal hydroxyl groups in the 1,3,5-triazinenucleus enhances the activity of some of the compounds(26, 27, 29, 32 and 33) towards both the strains (D6 andW2 clone) of P. falciparum with IC50 ranging from 0.10 to0.19 lM against D6 clone and 0.28–0.49 lM againstW2 clone. In this series (18–33), the change of piperidine(27–33) from morpholine group (18–26) improved the anti-malarial activity of these compounds against both thestrains of P. falciparum (IC50 ranging from 0.10 to 0.21 lMagainst D6 clone and 0.28–1.17 lM against W2 clone),while the increase in the carbon chain length of aliphaticaminoalcohols attached to 1,3,5-triazine nucleus generallyincreases the antimalarial activity towards W2 clone for aparticular linker (C2, C3 or C4) present between the ami-noquinoline and triazine moiety. Most of the compoundswere found to be non-cytotoxic up to the highest testedconcentration of 25 lM against all three cell lines (Table 2),indicating their safety in mammalian system. The mecha-nistic studies and structural modification of the lead mole-cules are under progress, and results will be published indue course of time.

Conclusion

In summary, systematic chemical modification of previouslyreported 4-aminoquinoline-triazine-based hybrids led tothe discovery of more potent 4-aminoquinoline-1,3,5-triazine-based hybrids with potent activity against bothCQ-sensitive (D6 clone) and CQ-resistant (W2 clone) ofP. falciparum. Ten compounds (6, 8, 10, 11, 13, 14, 16,27, 29 and 33) of total 28 compounds synthesised havedisplayed better activity than CQ against W2 clone ofP. falciparum. Introduction of aminoalcohol side chain withfree terminal hydroxyl group enhances the activity of com-pounds (26, 27, 29, 32 and 33) towards both the strainsof P. falciparum. The activity profile of these molecules, inparticular of compounds 16 and 29, can be beneficial forthe generation of future drug candidates in malarialchemotherapy.

Acknowledgments

DSR thanks University Grants Commission [F. No. 41-202/2012(SR)], New Delhi, India and University of Delhi, Delhi,India for financial support. SM is thankful to CSIR for theaward of senior research fellowship. Thanks to USIC-CIF,University of Delhi for analytical data. SIK is thankful toUnited States Department of Agriculture (USDA), Agricul-tural Research Service Specific Cooperative AgreementNo. 58-6408-2-0009 for partial support of this work. Theauthors declare no conflict of interest.

References

1. Kumar N., Singh R., Rawat D.S. (2012) Tetraoxanes:synthetic and medicinal chemistry perspective. MedRes Rev;32:581–610.

2. WHO (2009) World Malaria Report. WHO http://www.who.int/malaria/world_malaria_report_2009/en/index.html (accessed on 26 January 2013)..

3. Krogstad D.J. (1996) Malaria as a re-emerging disease.Epidemiol Rev;18:77–89.

4. Greenwood B.M., Bojang K., Whitty C.J., Targett G.A.(2005) Malaria. Lancet;365:1487–1498.

5. O’Neill P.M., Bray P.G., Hawley S.R., Ward S.A., ParkB.K. (1998) 4-Aminoquinolines–past, present, andfuture: a chemical perspective. Pharmacol Ther;77:29–58.

6. Martin S.K., Oduola A.M., Milhous W.K. (1987) Rever-sal of chloroquine resistance in Plasmodium falciparum

by verapamil. Science;235:899–901.7. Burgess S.J., Selzer A., Kelly J.X., Smilkstein M.J.,

Riscoe M.K., Peyton D.H. (2006) A chloroquine-likemolecule designed to reverse resistance in Plasmodium

falciparum. J Med Chem;49:5623–5625.8. Mutabingwa T.K. (2005) Artemisinin-based combination

therapies (ACTs): best hope for malaria treatment butinaccessible to the needy!. Acta Trop;95:305–315.

Table 2: In vitro cytotoxicity of 4-aminoquinoline-triazine conju-gates (6–33) to mammalian cells

Entry VERO LLC-PK11 HepG2

6 NC NC NC7 NC NC NC8 >49.5 9.9 33.29 NC NC NC

10 28.8 8.6 11.511 NC 24.3 31.812 NC NC NC13 NC NC NC14 NC NC NC15 NC NC NC16 NC 27.8 NC17 NC NC NC18 NC NC NC19 NC NC NC20 NC NC NC21 NC NC NC22 NC NC NC23 NC NC NC24 NC NC NC25 NC NC NC26 NC NC NC27 NC NC NC28 NC NC NC29 NC NC NC30 NC NC NC31 >51.5 27.4 28.432 NC NC NC33 NC NC 40.9Doxorubicin >9.1 1.3 1.1

NC – not cytotoxic upto 25 lM; Vero – monkey kidney fibroblasts;LLC-PK11 – pig kidney epithelial cells; HepG2 – human hepatomacells.

628 Chem Biol Drug Des 2013; 81: 625–630

Manohar et al.

9. Kumar N., Sharma M., Rawat D.S. (2011) Medicinalchemistry perspectives of trioxanes and tetraoxanes.Curr Med Chem;18:3889–3928.

10. Dong Y., Tang Y., Chollet J., Matile H., Wittlin S.,Charman S.A., Charman W.N. et al. (2006) Effect offunctional group polarity on the antimalarial activity ofspiro and dispiro-1,2,4-trioxolanes. Bioorg MedChem;14:6368–6382.

11. Posner G.H., Paik I.H., Chang W., Borstnik K.,Sinishtaj S., Rosenthal A.S., Shapiro T.A. (2007)Malaria-infected mice are cured by a single dose ofnovel artemisinin derivatives. J Med Chem;50:2516–2519.

12. Paik I.H., Xie S., Shapiro T.A., Labonte T., NarducciSarjeant A.A., Baege A.C., Posner G.H. (2006) Secondgeneration, orally active, antimalarial, artemisinin-derived trioxane dimers with high stability, efficacy, andanticancer activity. J Med Chem;49:2731–2734.

13. Vennerstrom L., Arbe-Barnes S., Brun R., CharmanS.A., Chiu F.C., Chollet J., Dong Y. et al. (2004) Identi-fication of an antimalarial synthetic trioxolane drugdevelopment candidate. Nature;430:900–904.

14. O’Neill P.M., Posner G.H. (2004) A medicinal chemistryperspective on artmeisinin and related endoperoxides.J Med Chem;47:2945–2964.

15. Jambou R., Legrand E., Niang M., Khim N., Lim P.,Volney B., Ekala M.T., Bouchier C., Esterre P., Fan-deur T., Mercereau-Puijalon O. (2005) Resistance ofPlasmodium falciparum field isolates to in-vitro arteme-ther and point mutations of the SERCA-type PfATPase6. Lancet;366:1960–1963.

16. Egan T.J. (2001) Structure-function relationships inchloroquine and related 4-aminoquinoline antimalarials.Mini Rev Med Chem;1:113–123.

17. Dominguez J.N. (2002) Chemotherapeutic agentsagainst malaria: what next after chloroquine? Curr TopMed Chem;2:1173–1185.

18. Delarue S., Girault S., Maes L., Debreu-Fontaine M.A.,Labaeid M., Grellier P., Sergheraert C. (2001) Synthe-sis and in vitro and in vivo antimalarial activity of new4-anilinoquinolines. J Med Chem;44:2827–2833.

19. O’Neill P.M., Willock D.J., Hawley S.R., Bray P.G.,Storr R.C., Ward S.A., Park B.K. (1997) Synthesis,antimalarial activity, and molecular modeling of tebuqu-ine analogues. J Med Chem;40:437–448.

20. Madrid P.B., Liou A.P., DeRisi J.L., Guy R.K. (2006)Incorporation of an intramolecular hydrogen-bondingmotif in the side chain of 4-aminoquinolines enhancesactivity against drug-resistant P. falciparum. J MedChem;49:4535–4543.

21. Riccio E.S., Lee P.S., Winegar R.A., Krogstad D.J., DeD., Mirsalis J.C. (2001) Genetic toxicology testing ofthe antimalarial drugs chloroquine and a new analog,AQ-13. Environ Mol Mutagen;38:69–79.

22. Ridley R.G., Hofheinz H., Matile H., Jacquet C., DornA., Masciadri R., Jolidon S., Richter W.F., Guenzi A.,Girometta M.A., Urwyler H., Huber W., Thiathong S.,Peters W. (1996) 4-Aminoquinoline analogues of CQ

with shortened side chains retain activity against CQ-resistant Plasmodium falciparum. Antimicrob AgentsChemother;40:1846–1854.

23. De D., Krogstad F.M., Byers L.D., Krogstad D.J.(1998) Structure-activity relationships for antiplasmodialactivity among 7-substituted 4-aminoquinolines. J MedChem;41:4918–4926.

24. Hawley S.R., Bray P.G., Park B.K., Ward S.A. (1996)Amodiaquine accumulation in Plasmodium falciparum

as a possible explanation for its superior antimalarialactivity over chloroquine. Mol Biochem Parasitol;80:15–25.

25. Stocks P.A., Raynes K.J., Bray P.G., Park B.K., O’NeillP.M., Ward S.A. (2002) Novel short chain chloroquineanalogues retain activity against chloroquine resistantK1 Plasmodium falciparum. J Med Chem;45:4975–4983.

26. Madrid P.B., Wilson N.T., DeRisi J.L., Guy R.K. (2004)Parallel synthesis and antimalarial screening of a4-aminoquinoline library. J Comb Chem;6:437–442.

27. Dechy-Cabaret O., Benoit-Vical F., Robert A., MeunierB. (2000) Preparation and antimalarial activities of“trioxaquines”, new modular molecules with a trioxaneskeleton linked to a 4-aminoquinoline. ChemBio-Chem;1:281–283.

28. Peric M., Fajdetic A., Rupc R., Alihodzic S., Ziher D.,Krajacic M.B., Smith K.S. et al. (2012) Antimalarialactivity of 9a-n substituted 15-membered azalides withimproved in vitro and in vivo activity over azithromycin.J Med Chem;55:1389–1401.

29. Singh C., Malik H., Puri S.K. (2004) Synthesis and anti-malarial activity of a new series of trioxaquines. BioorgMed Chem;12:1177–1182.

30. Chiyanzu I., Clarkson C., Smith P.J., Lehman J., GutJ., Rosenthal P.J., Chibale K. (2005) Design, synthesisand anti-plasmodial evaluation in vitro of new 4-amino-quinoline isatin derivatives. Bioorg Med Chem;13:3249–3261.

31. Biot C., Glorian G., Maciejewski L.A., Brocard J.S.(1997) Synthesis and antimalarial activity in vitro and invivo of a new ferrocene-chloroquine analog. J MedChem;40:3715–3718.

32. Manohar S., Khan S.I., Rawat D.S. (2010) Synthesis,antimalarial activity and cytotoxicity of 4-aminoquino-line-triazine conjugates. Bioorg Med Chem Lett;20:322–325.

33. Manohar S., Khan S.I., Rawat D.S. (2011) Synthesis of4-aminoquinoline-1,2,3-triazole and 4-aminoquinoline-1,2,3-triazole-1,3,5-triazine hybrids as potential antima-larial agents. Chem Biol Drug Des;78:124–136.

34. Manohar S., Rajesh U.C., Khan S.I., Tekwani B.L.,Rawat D.S. (2012) Novel 4-aminoquinoline-pyrimidinebased hybrids with improved in vitro and in vivo anti-malarial activity. ACS Med Chem Lett;3:555–559.

35. Wenzel N.I., Chavain N., Wang Y., Friebolin W., MaesL., Pradines B., Lanzer M., Yardley V., Brun R., Her-old-Mende C., Biot C., Toth K., Davioud-Charvet E.(2010) Antimalarial versus cytotoxic properties of dual

Chem Biol Drug Des 2013; 81: 625–630 629

4-Aminoquinoline-triazine Based Hybrids

drugs derived from 4-aminoquinolines and mannichbases: interaction with DNA. J Med Chem;53:3214–3226.

36. Lombard M.C., N’Da D.D., Breytenbach J.C.,Smith P.J., Lategan C.A. (2011) Synthesis, in vitroantimalarial and cytotoxicity of artemisinin-amino-quinoline hybrids. Bioorg Med Chem Lett;21:1683–1686.

37. Sunduru N., Sharma M., Srivastava K., Rajakumar S.,Puri S.K., Saxena J.K., Chauhan P.M.S. (2009)Synthesis of oxalamide and triazine derivatives as anovel class of hybrid 4-aminoquinoline with potentantiplasmodial activity. Bioorg Med Chem;17:6451–6462.

38. Melato S., Prosperi D., Coghi P., Basilico N., Monti D.(2008) A combinatorial approach to 2,4,6-trisubstitutedtriazines with potent antimalarial activity: combiningconventional synthesis and microwave-assistance.ChemMedChem;3:873–876.

39. Atheaya H., Khan S.I., Mamgain R., Rawat D.S. (2008)Synthesis, thermal stability, antimalarial activity of sym-

metrically and asymmetrically substituted tetraoxanes.Bioorg Med Chem Lett;18:1446–1449.

40. Kumar N., Khan S.I., Beena, Rajalakshmi G., Kumara-dhas P., Rawat D.S. (2009) Synthesis, antimalarial activ-ity and cytotoxicity of substituted 3,6-diphenyl-[1,2,4,5]tetraoxanes. Bioorg Med Chem;17:5632–5638.

41. Kumar N., Khan S.I., Sharma M., Atheaya H., RawatD.S. (2009) Iodine-catalyzed one-pot synthesis andantimalarial activity evaluation of symmetrically andasymmetrically substituted 3,6-diphenyl[1,2,4,5]tetraox-anes. Bioorg Med Chem Lett;19:1675–1677.

42. Kumar N., Khan S.I., Atheaya H., Mamgain R., RawatD.S. (2011) Synthesis and in vitro antimalarial activityof tetraoxane-amine/amide conjugates. Eur J MedChem;46:2816–2827.

43. Natarajan J.K., Alumasa J.N., Yearick K., Ekoue-KoviK.A., Casabianca L.B., de Dios A.C., Wolf C., RoepeP.D. (2008) 4-N-, 4-S-, and 4-O-chloroquine ana-logues: influence of side chain length and quinolylnitrogen pKa on activity vs chloroquine resistantmalaria. J Med Chem;51:3466–3479.

630 Chem Biol Drug Des 2013; 81: 625–630

Manohar et al.