Natural Products and Bioprospecting    2023, Vol. 13 Issue (5) : 37-37     DOI: 10.1007/s13659-023-00402-2
ORIGINAL ARTICLES |
Prioritised identification of structural classes of natural products from higher plants in the expedition of antimalarial drug discovery
Phanankosi Moyo1, Luke Invernizzi1, Sephora M. Mianda1, Wiehan Rudolph1, Andrew W. Andayi2, Mingxun Wang3, Neil R. Crouch4,5, Vinesh J. Maharaj1
1. Department of Chemistry, Faculty of Natural and Agricultural Sciences, Biodiscovery Center, University of Pretoria, Private Bag X 20, Hatfield, Pretoria, 0028, South Africa;
2. Department of Physical and Biological Sciences, Murang'a University of Technology Murang'a, Murang'a, Kenya;
3. Computer Science and Engineering, University of California Riverside, 900 University Ave, Riverside, CA, 92521, USA;
4. Biodiversity Research and Monitoring Directorate, South African National Biodiversity Institute, Berea Road, P. O. Box 52099, Durban, 4007, South Africa;
5. School of Chemistry and Physics, University of KwaZulu-Natal, Durban, 4041, South Africa
Download: PDF(8149 KB)   HTML ()  
Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks    
Abstract  The emergence and spread of drug-recalcitrant Plasmodium falciparum parasites threaten to reverse the gains made in the fight against malaria. Urgent measures need to be taken to curb this impending challenge. The higher plant-derived sesquiterpene, quinoline alkaloids, and naphthoquinone natural product classes of compounds have previously served as phenomenal chemical scaffolds from which integral antimalarial drugs were developed. Historical successes serve as an inspiration for the continued investigation of plant-derived natural products compounds in search of novel molecular templates from which new antimalarial drugs could be developed. The aim of this study was to identify potential chemical scaffolds for malaria drug discovery following analysis of historical data on phytochemicals screened in vitro against P. falciparum. To identify these novel scaffolds, we queried an in-house manually curated database of plant-derived natural product compounds and their in vitro biological data. Natural products were assigned to different structural classes using NPClassifier. To identify the most promising chemical scaffolds, we then correlated natural compound class with bioactivity and other data, namely (i) potency, (ii) resistance index, (iii) selectivity index and (iv) physicochemical properties. We used an unbiased scoring system to rank the different natural product classes based on the assessment of their bioactivity data. From this analysis we identified the top-ranked natural product pathway as the alkaloids. The top three ranked super classes identified were (i) pseudoalkaloids, (ii) naphthalenes and (iii) tyrosine alkaloids and the top five ranked classes (i) quassinoids (of super class triterpenoids), (ii) steroidal alkaloids (of super class pseudoalkaloids) (iii) cycloeudesmane sesquiterpenoids (of super class triterpenoids) (iv) isoquinoline alkaloids (of super class tyrosine alkaloids) and (v) naphthoquinones (of super class naphthalenes). Launched chemical space of these identified classes of compounds was, by and large, distinct from that of ‘legacy’ antimalarial drugs. Our study was able to identify chemical scaffolds with acceptable biological properties that are structurally different from current and previously used antimalarial drugs. These molecules have the potential to be developed into new antimalarial drugs.
Keywords Natural products      Compound classes      Phytochemicals      Plasmodium falciparum      Antiplasmodial drug resistance      Malaria      Drug development     
Fund:This research work was supported by a grant from the Department of Science and Innovation (DSI) of South Africa awarded to Vinesh Maharaj. The UP ISMC acknowledges the South African Medical Research Council as Collaborating Centre for Malaria Research. Phanankosi Moyo was supported by a grant from the University of Pretoria Postgraduate Research Support Bursary for his Postdoctoral Fellowship. Sephora Mianda Mutombo was supported by funds from the University of Pretoria Postgraduate Research Support Bursary, South Africa and the L’Oréal-UNESCO for Woman in Science grant. Luke Invernizzi was supported by funds from the National Research Foundation of South Africa.
Corresponding Authors: Vinesh J. Maharaj,E-mail:vinesh.maharaj@up.ac.za     E-mail: vinesh.maharaj@up.ac.za
Issue Date: 03 November 2023
Service
E-mail this article
E-mail Alert
RSS
Articles by authors
Phanankosi Moyo
Luke Invernizzi
Sephora M. Mianda
Wiehan Rudolph
Andrew W. Andayi
Mingxun Wang
Neil R. Crouch
Vinesh J. Maharaj
Trendmd:   
Cite this article:   
Phanankosi Moyo,Luke Invernizzi,Sephora M. Mianda, et al. Prioritised identification of structural classes of natural products from higher plants in the expedition of antimalarial drug discovery[J]. Natural Products and Bioprospecting, 2023, 13(5): 37-37.
URL:  
http://npb.kib.ac.cn/EN/10.1007/s13659-023-00402-2     OR     http://npb.kib.ac.cn/EN/Y2023/V13/I5/37
[1] WHO, World malaria report 2022. 2022: World Health Organization.
[2] Menard D, Dondorp A. Antimalarial drug resistance: a threat to malaria elimination. Cold Spring Harb Perspect Med. 2017;7(7): a025619.
[3] Takala-Harrison S, et al. Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis. 2015;211(5):670-9.
[4] Wells TNC. Natural products as starting points for future anti-malarial therapies: going back to our roots? Malar J. 2011;10(1):S3.
[5] Gaillard T, et al. Antibiotics in malaria therapy: which antibiotics except tetracyclines and macrolides may be used against malaria? Malar J. 2016;15(1):556.
[6] Organization WH. Guidelines for the treatment of malaria. 2015: World Health Organization.
[7] Spížek J, Řezanka T. Lincomycin, clindamycin and their applications. Appl Microbiol Biotechnol. 2004;64(4):455-64.
[8] Nelson ML, Levy SB. The history of the tetracyclines. Ann N Y Acad Sci. 2011;1241(1):17-32.
[9] Peters W. The evolution of tafenoquine—antimalarial for a new millennium? J R Soc Med. 1999;92(7):345-52.
[10] Watson JA, Nekkab N, White M. Tafenoquine for the prevention of Plasmodium vivax malaria relapse. Lancet Microbe. 2021;2(5):e175-6.
[11] Wang M, et al. Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking. Nat Biotechnol. 2016;34(8):828-37.
[12] Dührkop K, et al. SIRIUS 4: a rapid tool for turning tandem mass spectra into metabolite structure information. Nat Methods. 2019;16(4):299-302.
[13] Chassagne F, et al. The landscape of natural product diversity and their pharmacological relevance from a focus on the Dictionary of Natural Products®. Phytochem Rev. 2019;18:601-22.
[14] Egieyeh SA, et al. Prioritization of anti-malarial hits from nature: chemo-informatic profiling of natural products with in vitro antiplasmodial activities and currently registered anti-malarial drugs. Malar J. 2016;15:1-23.
[15] Kim HW, et al. NPClassifier: a deep neural network-based structural classification tool for natural products. J Nat Prod. 2021;84(11):2795-807.
[16] Djoumbou Feunang Y, et al. ClassyFire: automated chemical classification with a comprehensive, computable taxonomy. J Cheminformat. 2016;8:1-20.
[17] Hai Y, et al. Trends of antimalarial marine natural products: progresses, challenges and opportunities. Nat Prod Rep. 2022;39(5):969-90.
[18] Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7(1):42717.
[19] Lipinski CA, et al. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 1997;23(1):3-25.
[20] Veber DF, et al. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem. 2002;45(12):2615-23.
[21] Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J Comb Chem. 1999;1(1):55-68.
[22] Chakraborty D, Pal A. Quassinoids: chemistry and novel detection techniques. Berlin: Springer; 2013. p. 3345-66.
[23] Duan Z-K, et al. Quassinoids: phytochemistry and antitumor prospect. Phytochemistry. 2021;187: 112769.
[24] Kraus GA, Taschner MJ. Model studies for the synthesis of quassinoids. 1. Construction of the BCE ring system. J Org Chem. 1980;45(6):1175-6.
[25] Herscovici J, et al. Stereocontrolled routes to functionalized [1,8-bc]naphthopyran. A study on the total synthesis of quassinoids and tetrahydronaphthalene antibiotics. J Org Chem. 1993;58(15):3928-37.
[26] Gross RS, Grieco PA, Collins JL. Synthetic studies on quassinoids: total synthesis of (±)-chaparrinone. J Am Chem Soc. 1990;112(25):9436-7.
[27] Ziegler FE, et al. Practical routes to two functionalized decalones for the synthesis of quassinoids. J Org Chem. 1986;51(24):4573-9.
[28] Thomas WP, Pronin SV. A concise enantioselective approach to quassinoids. J Am Chem Soc. 2021;144(1):118-22.
[29] Pazur EJ, Wipf P. Recent syntheses and biological profiling of quassinoids. Org Biomol Chem. 2022;20(19):3870-89.
[30] Kawada K, Kim M, Watt DS. Synthesis of quassinoids. A review. Org Prep Proc Int. 1989;21(5):521-618.
[31] O’Neill MJ, et al. Plants as sources of antimalarial drugs: in vitro antimalarial activities of some quassinoids. Antimicrob Agents Chemother. 1986;30(1):101-4.
[32] O’Neill MJ, et al. Plants as sources of antimalarial drugs, Part 4: Activity of Brucea javanica fruits against chloroquine-resistant Plasmodium falciparum in vitro and against Plasmodium berghei in vivo. J Nat Prod. 1987;50(1):41-8.
[33] Tay DW, et al. 67 million natural product-like compound database generated via molecular language processing. Sci Data. 2023;10(1):296.
[34] Li Y, et al. Designing natural product-like virtual libraries using deep molecule generative models. Macromolecules. 2018;3:5.
[35] Yu MJ. Natural product-like virtual libraries: recursive atom-based enumeration. J Chem Inf Model. 2011;51(3):541-57.
[36] Abd Karim HA, Ismail NH, Osman CP. Steroidal alkaloids from the apocynaceae family: their isolation and biological activity. Nat Prod Commun. 2022;17(11):1934578X221141265.
[37] Xiang M-L, et al. Chemistry and bioactivities of natural steroidal alkaloids. Nat Prod Bioprospect. 2022;12(1):23.
[38] Sharpe RJ, Johnson JS. A global and local desymmetrization approach to the synthesis of steroidal alkaloids: stereocontrolled total synthesis of paspaline. J Am Chem Soc. 2015;137(15):4968-71.
[39] Tokuyama T, Daly J, Witkop B. Structure of batrachotoxin, a steroidal alkaloid from the Colombian arrow poison frog, Phyllobates aurotaenia, and partial synthesis of batrachotoxin and its analogs and homologs. J Am Chem Soc. 1969;91(14):3931-8.
[40] Zha X, et al. Efficient synthesis of solasodine, O-acetylsolasodine, and soladulcidine as anticancer steroidal alkaloids. Chem Biodivers. 2007;4(1):25-31.
[41] Szabó LU, et al. Antiprotozoal nor-triterpene alkaloids from Buxus sempervirens L. Antibiotics. 2021;10(6):696.
[42] Zhou B, et al. Nanomolar antimalarial agents against chloroquine-resistant Plasmodium falciparum from medicinal plants and their structure-activity relationships. J Nat Prod. 2017;80(1):96-107.
[43] Lombe BK, Feineis D, Bringmann G. Dimeric naphthylisoquinoline alkaloids: polyketide-derived axially chiral bioactive quateraryls. Nat Prod Rep. 2019;36(11):1513-45.
[44] Bringmann G, et al. Highly selective antiplasmodial naphthylisoquinoline alkaloids from Ancistrocladus tectorius. Phytochemistry. 2013;91:220-8.
[45] Bringmann G, et al. Habropetaline A, an antimalarial naphthylisoquinoline alkaloid from Triphyophyllum peltatum. Phytochemistry. 2003;62(3):345-9.
[46] Moyo P, et al. Naphthylisoquinoline alkaloids, validated as hit multistage antiplasmodial natural products. Int J Parasitol Drugs Drug Resist. 2020;13:51-8.
[47] Charman SA, et al. An in vitro toolbox to accelerate anti-malarial drug discovery and development. Malar J. 2020;19(1):1-27.
[48] Cihan Sorkun M, et al. ChemPlot, a Python library for chemical space visualization. Chem Methods. 2022;2(7): e202200005.
[1] Phanankosi Moyo, Luke Invernizzi, Sephora M. Mianda, Wiehan Rudolph, Warren A. Andayi, Mingxun Wang, Neil R. Crouch, Vinesh J. Maharaj. Leveraging off higher plant phylogenetic insights for antiplasmodial drug discovery[J]. Natural Products and Bioprospecting, 2023, 13(5): 35-35.
[2] Dalila Carbone, Carmela Gallo, Genoveffa Nuzzo, Giusi Barra, Mario Dell'Isola, Mario Affuso, Olimpia Follero, Federica Albiani, Clementina Sansone, Emiliano Manzo, Giuliana d'Ippolito, Angelo Fontana. Marine natural product lepadin A as a novel inducer of immunogenic cell death via CD91-dependent pathway[J]. Natural Products and Bioprospecting, 2023, 13(5): 34-34.
[3] Shah Faisal, Syed Lal Badshah, Bibi Kubra, Abdul, Hamid Emwas, and Mariusz Jaremko. Alkaloids as potential antivirals. A comprehensive review[J]. Natural Products and Bioprospecting, 2023, 13(1): 4-4.
[4] Ji-Kai Liu. Natural products in cosmetics[J]. Natural Products and Bioprospecting, 2022, 12(6): 40-40.
[5] Si-Yuan Luo, Jun-Yu Zhu, Ming-Feng Zou, Sheng Yin, Gui-Hua Tang. Mulberry Diels–Alder-type adducts: isolation, structure, bioactivity, and synthesis[J]. Natural Products and Bioprospecting, 2022, 12(5): 31-31.
[6] Ji-Kai Liu. Antiaging agents: safe interventions to slow aging and healthy life span extension[J]. Natural Products and Bioprospecting, 2022, 12(3): 18-18.
[7] Yulian Lv, Tian Tian, Yong-Jiang Wang, Jian-Ping Huang, Sheng-Xiong Huang. Advances in chemistry and bioactivity of the genus Erythroxylum[J]. Natural Products and Bioprospecting, 2022, 12(3): 15-15.
[8] Ghodsi Mohammadi Ziarani, Negar Jamasbi, Fatemeh Mohajer. Recent advances on the synthesis of natural pyrrolizidine alkaloids: alexine, and its stereoisomers[J]. Natural Products and Bioprospecting, 2022, 12(1): 1-15.
[9] Oyere Tanyi Ebob, Smith B. Babiaka, Fidele Ntie-Kang. Natural Products as Potential Lead Compounds for Drug Discovery Against SARS-CoV-2[J]. Natural Products and Bioprospecting, 2021, 11(6): 611-628.
[10] Christian Bailly, Gérard Vergoten. Anticancer Properties and Mechanism of Action of Oblongifolin C, Guttiferone K and Related Polyprenylated Acylphloroglucinols[J]. Natural Products and Bioprospecting, 2021, 11(6): 629-641.
[11] Patrick O. Sakyi, Richard K. Amewu, Robert N. O. A. Devine, Emahi Ismaila, Whelton A. Miller, Samuel K. Kwofie. The Search for Putative Hits in Combating Leishmaniasis: The Contributions of Natural Products Over the Last Decade[J]. Natural Products and Bioprospecting, 2021, 11(5): 489-544.
[12] Darko Jenic, Helen Waller, Helen Collins, Clett Erridge. Reversal of Tetracycline Resistance by Cepharanthine, Cinchonidine, Ellagic Acid and Propyl Gallate in a Multi-drug Resistant Escherichia coli[J]. Natural Products and Bioprospecting, 2021, 11(3): 345-356.
[13] Christian Bailly. Anticancer Properties of Lobetyolin, an Essential Component of Radix Codonopsis (Dangshen)[J]. Natural Products and Bioprospecting, 2021, 11(2): 143-153.
[14] Min Huang, Jin-Jian Lu, Jian Ding. Natural Products in Cancer Therapy: Past, Present and Future[J]. Natural Products and Bioprospecting, 2021, 11(1): 5-13.
[15] Christian Bailly. Anticancer Activities and Mechanism of Action of Nagilactones, a Group of Terpenoid Lactones Isolated from Podocarpus Species[J]. Natural Products and Bioprospecting, 2020, 10(6): 367-375.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed