The resistome and genomic reconnaissance in the age of malaria elimination

ABSTRACT Malaria is an infectious disease caused by parasitic protozoa in the Plasmodium genus. A complete understanding of the biology of these parasites is challenging in view of their need to switch between the vertebrate and insect hosts. The parasites are also capable of becoming highly motile and of remaining dormant for decades, depending on the stage of their life cycle. Malaria elimination efforts have been implemented in several endemic countries, but the parasites have proven to be resilient. One of the major obstacles for malaria elimination is the development of antimalarial drug resistance. Ineffective treatment regimens will fail to remove the circulating parasites and to prevent the local transmission of the disease. Genomic epidemiology of malaria parasites has become a powerful tool to track emerging drug-resistant parasite populations almost in real time. Population-scale genomic data are instrumental in tracking the hidden pockets of Plasmodium in nationwide elimination efforts. However, genomic surveillance data can be useful in determining the threat only when combined with a thorough understanding of the malarial resistome – the genetic repertoires responsible for causing and potentiating drug resistance evolution. Even though long-term selection has been a standard method for drug target identification in laboratories, its implementation in large-scale exploration of the druggable space in Plasmodium falciparum, along with genome-editing technologies, have enabled mapping of the genetic repertoires that drive drug resistance. This Review presents examples of practical use and describes the latest technology to show the power of real-time genomic epidemiology in achieving malaria elimination. Summary: This Review discusses the challenges in malaria elimination and how implementation of national-scale genomic surveillance programmes in combination with resistome analyses could provide a powerful solution.

[1]  X. Su,et al.  Genome-wide association analysis identifies genetic loci associated with resistance to multiple antimalarials in Plasmodium falciparum from China-Myanmar border , 2016, Scientific Reports.

[2]  G. Dantas,et al.  Next-generation approaches to understand and combat the antibiotic resistome , 2017, Nature Reviews Microbiology.

[3]  I. Rollo ‘Daraprim’ Resistance in Experimental Malarial Infections , 1952, Nature.

[4]  James R. Brown,et al.  Thousands of chemical starting points for antimalarial lead identification , 2010, Nature.

[5]  Hongshen Ma,et al.  (+)-SJ733, a clinical candidate for malaria that acts through ATP4 to induce rapid host-mediated clearance of Plasmodium , 2014, Proceedings of the National Academy of Sciences.

[6]  David M. Shackleford,et al.  Antimalarial efficacy of MMV390048, an inhibitor of Plasmodium phosphatidylinositol 4-kinase , 2017, Science Translational Medicine.

[7]  T. Hien,et al.  Malaria , 2014, The Lancet.

[8]  J. Rayner,et al.  Uncovering the essential genes of the human malaria parasite Plasmodium falciparum by saturation mutagenesis , 2018, Science.

[9]  T. Wells,et al.  The antimalarial pipeline , 2018, Current opinion in pharmacology.

[10]  Taane G. Clark,et al.  A global network for investigating the genomic epidemiology of malaria , 2008, Nature.

[11]  M. Llinás,et al.  Specific Inhibition of the Bifunctional Farnesyl/Geranylgeranyl Diphosphate Synthase in Malaria Parasites via a New Small-Molecule Binding Site. , 2017, Cell chemical biology.

[12]  E. Winzeler,et al.  Exploration of the Plasmodium falciparum Resistome and Druggable Genome Reveals New Mechanisms of Drug Resistance and Antimalarial Targets , 2018, Microbiology insights.

[13]  W. Milhous,et al.  Molecular basis of differential resistance to cycloguanil and pyrimethamine in Plasmodium falciparum malaria. , 1990, Proceedings of the National Academy of Sciences of the United States of America.

[14]  Gregory M. Goldgof,et al.  A broad analysis of resistance development in the malaria parasite , 2016, Nature Communications.

[15]  P. Newton,et al.  A Major Genome Region Underlying Artemisinin Resistance in Malaria , 2012, Science.

[16]  M. Fraser,et al.  High-efficiency transformation of Plasmodium falciparum by the lepidopteran transposable element piggyBac. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[17]  Joseph K. Pickrell,et al.  The Genetics of Human Adaptation: Hard Sweeps, Soft Sweeps, and Polygenic Adaptation , 2010, Current Biology.

[18]  A. Saul,et al.  The dihydrofolate reductase domain of rodent malarias: point mutations and pyrimethamine resistance. , 1994, Molecular and biochemical parasitology.

[19]  K. Lindblade,et al.  A Historical Review of WHO Certification of Malaria Elimination. , 2019, Trends in parasitology.

[20]  G. Dennis Shanks,et al.  Supplemental Appendix A: 240 Studies Assessed for Inclusion Two-year Evaluation of Intermittent Preventive Treatment for Children (iptc) Combined with Timely Home Treatment for Malaria Control In , 2015 .

[21]  M. Whittaker,et al.  The challenge of artemisinin resistance can only be met by eliminating Plasmodium falciparum malaria across the Greater Mekong subregion , 2014, Malaria Journal.

[22]  Philipp W. Messer,et al.  Population genomics of rapid adaptation by soft selective sweeps. , 2013, Trends in ecology & evolution.

[23]  W. Trager,et al.  Chloroquine resistance produced in vitro in an African strain of human malaria. , 1978, Science.

[24]  C. MacPherson,et al.  Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system , 2014, Nature Biotechnology.

[25]  J. Burrows,et al.  New developments in anti-malarial target candidate and product profiles , 2017, Malaria Journal.

[26]  K. Harlos,et al.  Targeting Prolyl-tRNA Synthetase to Accelerate Drug Discovery against Malaria, Leishmaniasis, Toxoplasmosis, Cryptosporidiosis, and Coccidiosis. , 2017, Structure.

[27]  Thanat Chookajorn How to combat emerging artemisinin resistance: Lessons from “The Three Little Pigs” , 2018, PLoS pathogens.

[28]  N. White Counter Perspective: Artemisinin Resistance: Facts, Fears, and Fables , 2012, The American journal of tropical medicine and hygiene.

[29]  D. Conway,et al.  Plasmodium knowlesi malaria in humans is widely distributed and potentially life threatening. , 2008, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[30]  D. Wirth,et al.  In Vitro Resistance Selections for Plasmodium falciparum Dihydroorotate Dehydrogenase Inhibitors Give Mutants with Multiple Point Mutations in the Drug-binding Site and Altered Growth , 2014, The Journal of Biological Chemistry.

[31]  Min Zhang,et al.  Artemisinin resistance phenotypes and K13 inheritance in a Plasmodium falciparum cross and Aotus model , 2018, Proceedings of the National Academy of Sciences.

[32]  S. Lindquist,et al.  The cytoplasmic prolyl-tRNA synthetase of the malaria parasite is a dual-stage target of febrifugine and its analogs , 2015, Science Translational Medicine.

[33]  P. Wilairat,et al.  Plasmodium falciparum malaria: Convergent evolutionary trajectories towards delayed clearance following artemisinin treatment. , 2016, Medical hypotheses.

[34]  Pedro L. Alonso,et al.  Some Lessons for the Future from the Global Malaria Eradication Programme (1955–1969) , 2011, PLoS medicine.

[35]  R. Moreira,et al.  Primaquine revisited six decades after its discovery. , 2009, European journal of medicinal chemistry.

[36]  Gilean McVean,et al.  Genetic architecture of artemisinin-resistant Plasmodium falciparum , 2015, Nature Genetics.

[37]  David M. Shackleford,et al.  A Triazolopyrimidine-Based Dihydroorotate Dehydrogenase Inhibitor with Improved Drug-like Properties for Treatment and Prevention of Malaria. , 2016, ACS infectious diseases.

[38]  B. Bergmann,et al.  A genetic system to study Plasmodium falciparum protein function , 2017, Nature Methods.

[39]  John A. Tallarico,et al.  Selective and Specific Inhibition of the Plasmodium falciparum Lysyl-tRNA Synthetase by the Fungal Secondary Metabolite Cladosporin , 2012, Cell host & microbe.

[40]  P. Shaw,et al.  Tools for attenuation of gene expression in malaria parasites. , 2017, International journal for parasitology.

[41]  David W. Gray,et al.  A novel multiple-stage antimalarial agent that inhibits protein synthesis , 2015, Nature.

[42]  M. Llinás,et al.  Site-specific genome editing in Plasmodium falciparum using engineered zinc-finger nucleases , 2012, Nature Methods.

[43]  Bruce Russell,et al.  Spiroindolones, a Potent Compound Class for the Treatment of Malaria , 2010, Science.

[44]  D. Kwiatkowski,et al.  Spread of artemisinin resistance in Plasmodium falciparum malaria. , 2014, The New England journal of medicine.

[45]  B. Tekwani,et al.  Understanding the mechanisms for metabolism-linked hemolytic toxicity of primaquine against glucose 6-phosphate dehydrogenase deficient human erythrocytes: evaluation of eryptotic pathway. , 2012, Toxicology.

[46]  Gerard D. Wright The antibiotic resistome: the nexus of chemical and genetic diversity , 2007, Nature Reviews Microbiology.

[47]  K. Silamut,et al.  Artemisinin resistance in Plasmodium falciparum malaria. , 2009, The New England journal of medicine.

[48]  Xin-Zhuan Su,et al.  The discovery of artemisinin and the Nobel Prize in Physiology or Medicine , 2015, Science China Life Sciences.

[49]  Victoria C. Corey,et al.  Mapping the malaria parasite druggable genome by using in vitro evolution and chemogenomics , 2017, Science.

[50]  Manuel Llinás,et al.  Open-source discovery of chemical leads for next-generation chemoprotective antimalarials , 2018, Science.

[51]  Dana Carroll,et al.  Enhancing Gene Targeting with Designed Zinc Finger Nucleases , 2003, Science.

[52]  A. Cowman,et al.  Chromosomal rearrangements and point mutations in the DHFR-TS gene of Plasmodium chabaudi under antifolate selection. , 1990, Molecular and biochemical parasitology.

[53]  J. Rayner,et al.  Functional Profiling of a Plasmodium Genome Reveals an Abundance of Essential Genes , 2017, Cell.

[54]  P. Garner,et al.  Primaquine or other 8‐aminoquinolines for reducing Plasmodium falciparum transmission , 2018, The Cochrane database of systematic reviews.

[55]  B. Genton,et al.  A molecular marker of artemisinin-resistant Plasmodium falciparum malaria , 2013, Nature.

[56]  E. Crawford,et al.  Plasmid-free CRISPR/Cas9 genome editing in Plasmodium falciparum confirms mutations conferring resistance to the dihydroisoquinolone clinical candidate SJ733 , 2017, PLoS ONE.

[57]  Elizabeth A. Winzeler,et al.  Na+ Regulation in the Malaria Parasite Plasmodiumfalciparum Involves the Cation ATPase PfATP4 and Is a Target of the Spiroindolone Antimalarials , 2013, Cell host & microbe.

[58]  Anang A. Shelat,et al.  Chemical genetics of Plasmodium falciparum , 2010, Nature.

[59]  Thanat Chookajorn,et al.  "Snakes and Ladders" of drug resistance evolution , 2011, Virulence.

[60]  S. Hay,et al.  G6PD deficiency: global distribution, genetic variants and primaquine therapy. , 2013, Advances in parasitology.

[61]  Mehul Dhorda,et al.  The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study , 2017, The Lancet. Infectious diseases.

[62]  T. Kwan-Gett,et al.  Use cases for genetic epidemiology in malaria elimination , 2019, Malaria Journal.

[63]  Randall J. Platt,et al.  Efficient CRISPR/Cas9-mediated genome editing in P. falciparum , 2014, Nature Methods.

[64]  Robert W. Sauerwein,et al.  Targeting Plasmodium PI(4)K to eliminate malaria , 2013, Nature.

[65]  J. Derisi,et al.  Antimalarial Benzoxaboroles Target Plasmodium falciparum Leucyl-tRNA Synthetase , 2016, Antimicrobial Agents and Chemotherapy.

[66]  S. Schreiber,et al.  Synthesis of a Bicyclic Azetidine with In Vivo Antimalarial Activity Enabled by Stereospecific, Directed C(sp3)–H Arylation , 2017, Journal of the American Chemical Society.

[67]  Benito Munoz,et al.  Diversity-oriented synthesis yields novel multistage antimalarial inhibitors , 2016, Nature.

[68]  E. Winzeler,et al.  Using in Vitro Evolution and Whole Genome Analysis To Discover Next Generation Targets for Antimalarial Drug Discovery , 2018, ACS infectious diseases.

[69]  A. Nzila,et al.  In vitro selection of Plasmodium falciparum drug-resistant parasite lines , 2009, The Journal of antimicrobial chemotherapy.

[70]  Jim Stalker,et al.  Origins of the current outbreak of multidrug-resistant malaria in southeast Asia: a retrospective genetic study , 2017, bioRxiv.

[71]  N. White,et al.  Qinghaosu (Artemisinin): The Price of Success , 2008, Science.

[72]  D. Conway,et al.  Long read assemblies of geographically dispersed Plasmodium falciparum isolates reveal highly structured subtelomeres , 2018, Wellcome open research.

[73]  Gilean McVean,et al.  Multiple populations of artemisinin-resistant Plasmodium falciparum in Cambodia , 2013, Nature Genetics.

[74]  E. Ashley,et al.  Malaria , 2018, The Lancet.