Submit manuscript...
Journal of
eISSN: 2473-0831

Analytical & Pharmaceutical Research

Mini Review Volume 5 Issue 2

Antimalaria Drug Development & Pipeline

Lahiry Sandeep, Sinha Rajasree

Correspondence: Sandeep Lahiry, Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244 B A.J.C Bose Road, Kolkata, India

Received: April 25, 2017 | Published: June 22, 2017

Citation: Sandeep L, Rajasree S (2017) Antimalaria Drug Development & Pipeline. J Anal Pharm Res 5(2): 00136. DOI: 10.15406/japlr.2017.05.00136

Download PDF

Abstract

Increasing incidence of artemisin resistance endangers very foundations of current guideline based antimalarial therapy. There is an unmet need to develop newer strategies, targeting novel pathophysiology to set high standards in antimalaria care. Of late, the antimalarial drug pipeline is becoming increasingly robust, and promises healthier outcomes. We discuss few drugs currently under pre-clinical development that have shown encouraging results.

Keywords: Artemisin; Resistance; Malaria; Novel drugs

Abbreviations

Artemisinin-Based Combination Therapies; NCE: New Chemical Enitity; PfCRT: P. falciparum Chloroquine Resistance Transporter; IPT: Intermittent Preventative Treatment; MMV: Medicines for Malaria Ventures

Discussion

Artemisinin-based combination therapies (ACTs) are currently the gold standard treating uncomplicated malaria. Primaquine also eliminates liver hypnozoites, and is the only registered drug active against the mature gametocyte. However, resistance against existing antimalarials is well documented, and troubling due to the emerging resistance to artemisinins. A rising incidence of drug resistance requires new drugs with novel disease targeting strategies. The challenge is to demonstrate:

  1. Faster onset and longer duration of drug action,
  2. Safe for children and pregnant women and
  3. Ideally be amenable to a single-dose administration.

Artemisinin have traditionally cleave the peroxide bond by Fe(II) found in heme proteins, thus generating toxic oxygen radicals. Synthetic peroxides, thus are proving to be useful substitutes for artemisinin. The first-generation ozonide OZ277, known as arterolane [1], has been found to inhibit the growth of chloroquine-resistant (K1) and chloroquine sensitive (NF54) parasite strains. In 2012, the combination of arterolane maleate and piperaquine phosphate was released as a 3-day treatment in India [2]. The second-generation peroxide OZ439 (EC50 = 3.4–4.0nM) is now undergoing Phase IIa studies. It features an 80- aryl rather than an 80-alkyl group causing higher stability of the O– O bond towards Fe(II) increasing by 50-fold, presumably because of steric reasons. This in turn translates into a much longer half-life in both rats (t1/2 = 20 h for OZ439 vs. 1h for OZ277) and humans (t1/ 2 = 25–30h for OZ439) [3].

Tetraoxanes (also stabilizes O–O bond), has been employed in the drug candidate RKA 182, which has displayed good activity against P. falciparum 3D 7 strain and K1 strain (chloroquine sensitive and -resistant, respectively). [4] However, RKA 182 was not found curative in a single dose. The Central Drug Research Institute, Lucknow, India, is investigating a new chemical enitity (NCE) trioxane CDRI-97/78, currently in Phase I studies. It has ‘triaxone core ’and singlet oxygen to yield a peroxide compound [5].

P. falciparum chloroquine resistance transporter (PfCRT) mutations result in increased efflux of chloroquine from the acidic digestive vacuole to the cytosol of the parasite. Ferroquine has been found to be active against chloroquine- resistant strains, and is currently undergoing Phase II clinical trials. Ferroquine, unlike chloroquine, accumulates in the digestive vacuole of the chloroquine resistant parasites, enabling PfCRT inhibition [6]. Amodiaquine has been found active against most chloroquine resistant strains, however, two reactive metabolites are formed, namely imine and aldehyde, and are the likely causes of reported hepatotoxicity and agranulocytosis, respectively [7].

N-tert-Butyl isoquine (GSK369796) has been designed to avoid the formation of quinone imines, and has entered Phase I studies. It is potent in vitro, including in the chloroquine resistant strain K1 and is active in vivo, thus being comparable to amodiaquine. However, its development was discontinued due to exposures insufficient to demonstrate drug safety superior to chloroquine [8].

Walter Reed Army Institute of Research screened for analogs with a lower brain penetration, and have identified WR621308, which has a substantially lower permeability across MDCK cell monolayers than mefloquine, suggesting lower brain exposures [9].

Cycloguanil and pyrimethamine demonstrate inhibition of dihydrofolate reductase (DHFR). Inhibition of DHFR therefore arrests DNA replication, but resistance is widespread due to mutations in the enzyme [10]. P218, another DHFR inhibitor has been found to be active against all clinically relevant mutations. It combines the pyrimidine ring of pyrimethamine which brings potency, and the linker of the DHFR inhibitor WR99210, which tolerates mutations due to its flexibility. P218 is more potent than pyrimethamine against DHFR in the wild-type strain TM4 as well as in the quadruple mutant strain V1/S [11].

About 125 million pregnancies are at risk of malaria every year, and 10,000 women and 200,000 babies die as a result. An intermittent preventative treatment (IPT) has been recommended for pregnant women, but drug-resistance to the currently adopted IPT (sulfadoxine–pyrimethamine) poses an issue[12]. Azithromycin and chloroquine have demonstrated safety in children and pregnant women over a number of years. Notably, the azithromycin-chloroquine combination has been designed to be synergistic against chloroquine-resistant strains of P. falciparum, and was shown to be synergistic in the treatment of symptomatic malaria in clinical trials, with a maximum antiparasitic effect occurring only after two cycles of intra-erythrocytic development (one cycle of invasion, development, and egress lasts 42-48h). Finding azithromycin analogs with improved activity in mouse models of malaria has been challenging [13].

Spiroindolones as a novel chemotype series, has been optimized to deliver NITD-609, are now currently in Phase II trials [14]. The target was identified to be the cation channel PfATPase4. NITD-609 has an excellent potency, with 100% orally bioavailability in mouse and rat. It is also a potent inhibitor of gameto-cytogenesis, and blocks transmission to mosquitoes. The Medicines for Malaria Ventures (MMV) selected the spiroindolone project as the ‘Project of the Year 2009’ [15].

Albitiazolium (T3 or SAR97276) is a drug that has reached Phase II clinical trials. It acts primarily by inhibiting the transport of choline into the parasite. An important property of albitiazolium is that it accumulates irreversibly in the Plasmodium up to 1000-fold. Albitiazolium inhibits parasite growth and halts disease progression in mice without recrudescence [16]. DSM265 (Phase I) inhibits Pf DHODH (Dihydroo rotate dehydrogenase (DHODH) is the enzyme which catalyzes the rate-limiting step of the de novo pyrimidine biosynthetic pathway) selectively over its human counterpart. It demonstrated good oral bioavailability in rats and was efficacious in vitro and in mouse [17]. Benzimidazole inhibiting PfDHODH (IC50 = 40nM) and parasite growth, has a decent bioavailability in rat (49%) [18].

Genz-668764 inhibits P. falciparum in vitro and is active in mouse at doses of the order of 100mg/kg/day [19]. It is also active against the chloroquine resistant strain Dd2. Similarly, ML238 has been a potent NCE, being highly water soluble and not cytotoxic [20].

ACT-213615 has been established as fast-acting molecule against all asexual erythrocytic stages, currently being tested with encouraging results [21]. Notably, ACT-213615 completely cured P. berghei-infected mice with three consecutive oral daily doses of 750mg/kg. Benzoxaborole has also emerged as a promising starting agent [22]. TDR84420 was identified as a potent screening hit [WHO Special Programme for Research and Training in Tropical Diseases (TDR)] [23].

Conclusion

There have been significant advancements in antimalarials drug development in the preclinical setting, with few molecules showing exceptional properties. It will be interesting to see how many of these replicate such results in human trials.

References

  1. Dong Y, Wittlin S, Sriraghavan K, Chollet J, Charman SA, et al. (2009) The structure-activity relationship of the antimalarial ozonide Arterolane (OZ277). Journal of medicinal chemistry 53(1): 481-491.
  2. Gautam A, Ahmed T, Sharma P, Varshney B, Kothari M, et al. (2011) Pharmacokinetics and pharmacodynamics of arterolane maleate following multiple oral doses in adult patients with P. falciparum malaria. J Clin Pharmaco 51(11): 1519-1528.
  3. Charman SA, Arbe-Barnes S, Bathurst IC, Brun R, Campbell M, et al. (2011) Synthetic ozonide drug candidate OZ439 offers new hope for a single-dose cure of uncomplicated malaria. Proc Natl Acad Sci USA 108(11): 4400-4405.
  4. O'Neill PM, Amewu RK, Nixon GL, Bousejra ElGarah F, Mathirut M, et al. (2010) Identification of a 1, 2, 4, 5‐Tetraoxane Antimalarial Drug‐Development Candidate (RKA 182) with Superior Properties to the Semisynthetic Artemisinins. Angewandte chemie international edition 49(33): 5693-5697.
  5. Shafiq N, Rajagopalan S, Kushwaha HN, Mittal N, Chandurkar N, et al. (2014) Single ascending dose safety and pharmacokinetics of CDRI-97/78: first-in-human study of a novel antimalarial drug. Malar Res Treat 2014:372521.
  6. Biot C (2004) Ferroquine: a new weapon in the fight against malaria. Current Medicinal Chemistry-Anti-Infective Agents 3(2): 135-147.
  7. Olliaro PC, Nevill C, LeBras J, Ringwald P, Mussano P, et al. (1996) Systematic review of amodiaquine treatment in uncomplicated malaria. Lancet 348(9036): 1196-1201.
  8. O’Neill PM, Park BK, Shone AE, Maggs JL, Roberts P, et al. (2009) Candidate selection and preclinical evaluation of N-tert-butyl isoquine (GSK369796), an affordable and effective 4-aminoquinoline antimalarial for the 21st century. J Med Chem 52(5): 1408-1415.
  9. Milner E, Gardner S, Moon J, Grauer K, Auschwitz J, et al. (2011) Structure–activity relationships of 4-position diamine quinoline methanols as intermittent preventative treatment (IPT) against Plasmodium falciparum. J Med Chem 54(18): 6277-6285.
  10. Costanzo MS, Brown KM, Hartl DL (2011) Fitness trade-offs in the evolution of dihydrofolate reductase and drug resistance in Plasmodium falciparum. PLoS One 6: e19636.
  11. Yuthavong Y, Tarnchompoo B, Vilaivan T, Chitnumsub P, Kamchonwongpaisan S, et al. (2012) Malarial dihydrofolate reductase as a paradigm for drug development against a resistance-compromised target. Proc Natl Acad Sci USA 109(42): 16823-16828.
  12. ter Kuile FO, van Eijk AM, Filler SJ (2007) Effect of sulfadoxine-pyrimethamine resistance on the efficacy of intermittent preventive therapy for malaria control during pregnancy: a systematic review. JAMA 297(23): 2603-2616.
  13. Chico RM, Pittrof R, Greenwood B, Chandramohan D (2008) Azithromycin-chloroquine and the intermittent preventive treatment of malaria in pregnancy. Malar J 7: 255.
  14. Astelbauer F, Walochnik J (2011) Antiprotozoal compounds: state of the art and new developments. Int J Antimicrob Agents 38(2): 118-124.
  15. Rottmann M, McNamara C, Yeung BK, Lee MC, Zou B, et al. (2010) Spiroindolones, a potent compound class for the treatment of malaria. Science 329(5996): 1175-1180.
  16. Caldarelli SA, Hamel M, Duckert JF, Ouattara M, Calas M, et al. (2012) Disulfide prodrugs of albitiazolium (T3/SAR97276): synthesis and biological activities. J Med Chem 55(10): 4619-4628.
  17. Phillips MA, Lotharius J, Marsh K, White J, Dayan A, et al. (2015) A long-duration dihydroorotate dehydrogenase inhibitor (DSM265) for prevention and treatment of malaria. Sci Transl Med 7(296): 296ra111.
  18. Worachartcheewan A, Nantasenamat C, Isarankura-Na-Ayudhya C, Prachayasittikul V (2013) QSAR study of amidino bis-benzimidazole derivatives as potent anti-malarial agents against Plasmodium falciparum. Chemical Papers 67(11): 1462-1473.
  19. Younis Y, Street LJ, Waterson D, Witty MJ, Chibale K (2013) Cell-Based Medicinal Chemistry Optimization of High Throughput Screening Hits for Orally Active Antimalarials. Part 2: Hits from Soft Focus Kinase and other Libraries: Miniperspectives Series on Phenotypic Screening for Antiinfective Targets. Journal of medicinal chemistry 56(20): 7750-7754.
  20. Weiwer M, Mulrooney C, Massi D, Heidebrecht R, Wiegand R, et al. (2011) ML238: an antimalarial small molecule of a unique structural class.
  21. Brunner R, Ng CL, Aissaoui H, Akabas MH, Boss C, et al. (2013) UV-triggered affinity capture identifies interactions between the Plasmodium falciparum multidrug resistance protein 1 (PfMDR1) and antimalarial agents in live parasitized cells. J Biol Chem 288(31): 22576-22583.
  22. Zhang YK, Plattner JJ, Easom EE, Waterson D, Ge M, et al. (2011) An efficient synthesis for a new class antimalarial agent, 7-(2-carboxyethyl)-1, 3-dihydro-1-hydroxy-2, 1-benzoxaborole. Tetrahedron Letters 52(30): 3909-3911.
  23. Biamonte MA, Wanner J, Le Roch KG (2013) Recent advances in malaria drug discovery. Bioorganic & medicinal chemistry letters 23(10): 2829-2843.
Creative Commons Attribution License

©2017 Sandeep, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.