Targeting malaria parasites with novel derivatives of azithromycin

Introduction The spread of artemisinin resistant Plasmodium falciparum parasites is of global concern and highlights the need to identify new antimalarials for future treatments. Azithromycin, a macrolide antibiotic used clinically against malaria, kills parasites via two mechanisms: ‘delayed death’ by inhibiting the bacterium-like ribosomes of the apicoplast, and ‘quick-killing’ that kills rapidly across the entire blood stage development. Methods Here, 22 azithromycin analogues were explored for delayed death and quick-killing activities against P. falciparum (the most virulent human malaria) and P. knowlesi (a monkey parasite that frequently infects humans). Results Seventeen analogues showed improved quick-killing against both Plasmodium species, with up to 38 to 20-fold higher potency over azithromycin after less than 48 or 28 hours of treatment for P. falciparum and P. knowlesi, respectively. Quick-killing analogues maintained activity throughout the blood stage lifecycle, including ring stages of P. falciparum parasites (<12 hrs treatment) and were >5-fold more selective against P. falciparum than human cells. Isopentenyl pyrophosphate supplemented parasites that lacked an apicoplast were equally sensitive to quick-killing analogues, confirming that the quick killing activity of these drugs was not directed at the apicoplast. Further, activity against the related apicoplast containing parasite Toxoplasma gondii and the gram-positive bacterium Streptococcus pneumoniae did not show improvement over azithromycin, highlighting the specific improvement in antimalarial quick-killing activity. Metabolomic profiling of parasites subjected to the most potent compound showed a build-up of non-haemoglobin derived peptides that was similar to chloroquine, while also exhibiting accumulation of haemoglobin-derived peptides that was absent for chloroquine treatment. Discussion The azithromycin analogues characterised in this study expand the structural diversity over previously reported quick-killing compounds and provide new starting points to develop azithromycin analogues with quick-killing antimalarial activity.

[1]  T. Horii,et al.  Evidence of Artemisinin-Resistant Malaria in Africa. , 2021, The New England journal of medicine.

[2]  S. Prigge,et al.  Dephospho‐CoA kinase, a nuclear‐encoded apicoplast protein, remains active and essential after Plasmodium falciparum apicoplast disruption , 2021, The EMBO journal.

[3]  A. Dicko,et al.  The Duration of Protection from Azithromycin Against Malaria, Acute Respiratory, Gastrointestinal, and Skin Infections When Given Alongside Seasonal Malaria Chemoprevention: Secondary Analyses of Data from a Clinical Trial in Houndé, Burkina Faso, and Bougouni, Mali , 2021, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[4]  F. Gamo,et al.  A novel class of fast‐acting antimalarial agents: Substituted 15‐membered azalides , 2020, British journal of pharmacology.

[5]  Danny W. Wilson,et al.  Retargeting azithromycin analogues to have dual-modality antimalarial activity , 2020, BMC Biology.

[6]  Shane M. Devine,et al.  Multi-omic Characterization of the Mode of Action of a Potent New Antimalarial Compound, JPC-3210, Against Plasmodium falciparum* , 2019, Molecular & Cellular Proteomics.

[7]  E. Winzeler,et al.  Combining Stage Specificity and Metabolomic Profiling to Advance Antimalarial Drug Discovery , 2019, Cell chemical biology.

[8]  D. Kwiatkowski,et al.  Emergence of artemisinin-resistant Plasmodium falciparum with kelch13 C580Y mutations on the island of New Guinea , 2019, bioRxiv.

[9]  S. Ralph,et al.  Delayed death in the malaria parasite Plasmodium falciparum is caused by disruption of prenylation-dependent intracellular trafficking , 2019, PLoS biology.

[10]  A. Dicko,et al.  Effect of Adding Azithromycin to Seasonal Malaria Chemoprevention. , 2019, The New England journal of medicine.

[11]  S. Sørensen,et al.  Short- and long-term impacts of azithromycin treatment on the gut microbiota in children: A double-blind, randomized, placebo-controlled trial , 2018, EBioMedicine.

[12]  L. Cui,et al.  Randomized, Double-Blind, Placebo-Controlled Studies to Assess Safety and Prophylactic Efficacy of Naphthoquine-Azithromycin Combination for Malaria Prophylaxis in Southeast Asia , 2018, Antimicrobial Agents and Chemotherapy.

[13]  Danny W. Wilson,et al.  Cellular dissection of malaria parasite invasion of human erythrocytes using viable Plasmodium knowlesi merozoites , 2018, Scientific Reports.

[14]  David S. Wishart,et al.  MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis , 2018, Nucleic Acids Res..

[15]  G. McFadden,et al.  Validation of Putative Apicoplast-Targeting Drugs Using a Chemical Supplementation Assay in Cultured Human Malaria Parasites , 2017, Antimicrobial Agents and Chemotherapy.

[16]  V. Avery,et al.  Plasmodium falciparum in vitro continuous culture conditions: A comparison of parasite susceptibility and tolerance to anti-malarial drugs throughout the asexual intra-erythrocytic life cycle , 2017, International journal for parasitology. Drugs and drug resistance.

[17]  H. Venter,et al.  Synthesis and antibacterial activity of novel 3-O-descladinosylazithromycin derivatives. , 2017, European journal of medicinal chemistry.

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

[19]  S. Ralph,et al.  Metabolomics-Based Screening of the Malaria Box Reveals both Novel and Established Mechanisms of Action , 2016, Antimicrobial Agents and Chemotherapy.

[20]  P. Rosenthal Azithromycin for Malaria? , 2016, The American journal of tropical medicine and hygiene.

[21]  L. Tilley,et al.  Haemoglobin degradation underpins the sensitivity of early ring stage Plasmodium falciparum to artemisinins , 2016, Journal of Cell Science.

[22]  D. Stephens,et al.  Macrolide Resistance in Streptococcus pneumoniae , 2016, Front. Cell. Infect. Microbiol..

[23]  Danny W. Wilson,et al.  Macrolides rapidly inhibit red blood cell invasion by the human malaria parasite, Plasmodium falciparum , 2015, BMC Biology.

[24]  C. Menéndez,et al.  Sulphadoxine-pyrimethamine plus azithromycin for the prevention of low birthweight in Papua New Guinea: a randomised controlled trial , 2015, BMC Medicine.

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

[26]  J. Dumler,et al.  Short-Term Malaria Reduction by Single-Dose Azithromycin during Mass Drug Administration for Trachoma, Tanzania , 2014, Emerging infectious diseases.

[27]  R. Leak,et al.  Viability assays for cells in culture. , 2014, Journal of visualized experiments : JoVE.

[28]  Tiffany M. DeSimone,et al.  Expansion of host cellular niche can drive adaptation of a zoonotic malaria parasite to humans , 2013, Nature Communications.

[29]  Danny W. Wilson,et al.  Defining the Timing of Action of Antimalarial Drugs against Plasmodium falciparum , 2013, Antimicrobial Agents and Chemotherapy.

[30]  G. McFadden,et al.  Chemobiosynthesis of New Antimalarial Macrolides , 2012, Antimicrobial Agents and Chemotherapy.

[31]  Rainer Breitling,et al.  IDEOM: an Excel interface for analysis of LC-MS-based metabolomics data , 2012, Bioinform..

[32]  J. Vidal,et al.  Design, synthesis, and in vitro activity of novel 2'-O-substituted 15-membered azalides. , 2012, Journal of medicinal chemistry.

[33]  S. Alihodžić,et al.  Novel hybrid molecules based on 15-membered azalide as potential antimalarial agents. , 2012, European journal of medicinal chemistry.

[34]  W. Milhous,et al.  Antimalarial activity of 9a-N substituted 15-membered azalides with improved in vitro and in vivo activity over azithromycin. , 2012, Journal of medicinal chemistry.

[35]  Joseph L. DeRisi,et al.  Chemical Rescue of Malaria Parasites Lacking an Apicoplast Defines Organelle Function in Blood-Stage Plasmodium falciparum , 2011, PLoS biology.

[36]  W. Milhous,et al.  Synthesis, structure-activity relationship, and antimalarial activity of ureas and thioureas of 15-membered azalides. , 2011, Journal of medicinal chemistry.

[37]  J. Boothroyd,et al.  Chemistry and biology of macrolide antiparasitic agents. , 2011, Journal of medicinal chemistry.

[38]  D. Terlouw,et al.  Azithromycin for treating uncomplicated malaria. , 2011, The Cochrane database of systematic reviews.

[39]  Sanja Koštrun,et al.  Modeling cellular pharmacokinetics of 14- and 15-membered macrolides with physicochemical properties. , 2011, Journal of medicinal chemistry.

[40]  Y. Cheung,et al.  Effect of repeated treatment of pregnant women with sulfadoxine-pyrimethamine and azithromycin on preterm delivery in Malawi: a randomized controlled trial. , 2010, The American journal of tropical medicine and hygiene.

[41]  Dave Richard,et al.  Complement receptor 1 is the host erythrocyte receptor for Plasmodium falciparum PfRh4 invasion ligand , 2010, Proceedings of the National Academy of Sciences.

[42]  J. Collins,et al.  Bacterial charity work leads to population-wide resistance , 2010, Nature.

[43]  Danny W. Wilson,et al.  Isolation of viable Plasmodium falciparum merozoites to define erythrocyte invasion events and advance vaccine and drug development , 2010, Proceedings of the National Academy of Sciences.

[44]  K. Matuschewski,et al.  Natural Immunization Against Malaria: Causal Prophylaxis with Antibiotics , 2010, Science Translational Medicine.

[45]  B. Rubin,et al.  Mechanisms of Action and Clinical Application of Macrolides as Immunomodulatory Medications , 2010, Clinical Microbiology Reviews.

[46]  Leann Tilley,et al.  Digestive-vacuole genesis and endocytic processes in the early intraerythrocytic stages of Plasmodium falciparum , 2010, Journal of Cell Science.

[47]  J. Mckim Building a Tiered Approach to In Vitro Predictive Toxicity Screening: A Focus on Assays with In Vivo Relevance , 2010, Combinatorial chemistry & high throughput screening.

[48]  Shutao Ma,et al.  Synthesis and antibacterial activity of novel 11,12-cyclic carbonate azithromycin 4″-O-carbamate derivatives , 2010, The Journal of Antibiotics.

[49]  Danny W. Wilson,et al.  Development of fluorescent Plasmodium falciparum for in vitro growth inhibition assays , 2010, Malaria Journal.

[50]  Yoshiya Tanaka,et al.  Suppressive effect of azithromycin on Plasmodium berghei mosquito stage development and apicoplast replication , 2010, Malaria Journal.

[51]  C. Whitty,et al.  Azithromycin plus artesunate versus artemether-lumefantrine for treatment of uncomplicated malaria in Tanzanian children: a randomized, controlled trial. , 2009, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[52]  Shutao Ma,et al.  Synthesis and antibacterial activity of novel 15-membered macrolide derivatives: 4''-carbamate, 11,12-cyclic carbonate-4''-carbamate and 11,4''-di-O-arylcarbamoyl analogs of azithromycin. , 2009, European journal of medicinal chemistry.

[53]  M. Grainger,et al.  Formation of the Food Vacuole in Plasmodium falciparum: A Potential Role for the 19 kDa Fragment of Merozoite Surface Protein 1 (MSP119) , 2008, PloS one.

[54]  R. Hancock,et al.  Agar and broth dilution methods to determine the minimal inhibitory concentration (MIC) of antimicrobial substances , 2008, Nature Protocols.

[55]  P. Rosenthal,et al.  Multiple Antibiotics Exert Delayed Effects against the Plasmodium falciparum Apicoplast , 2007, Antimicrobial Agents and Chemotherapy.

[56]  G. McFadden,et al.  The effects of anti-bacterials on the malaria parasite Plasmodium falciparum. , 2007, Molecular and biochemical parasitology.

[57]  D. Fidock,et al.  In Vitro Efficacy, Resistance Selection, and Structural Modeling Studies Implicate the Malarial Parasite Apicoplast as the Target of Azithromycin* , 2007, Journal of Biological Chemistry.

[58]  D. Conway,et al.  A large focus of naturally acquired Plasmodium knowlesi infections in human beings , 2004, The Lancet.

[59]  R. Zarivach,et al.  Structural basis for the antibiotic activity of ketolides and azalides. , 2003, Structure.

[60]  Catherine Li,et al.  High-Throughput Growth Assay for Toxoplasma gondii Using Yellow Fluorescent Protein , 2003, Antimicrobial Agents and Chemotherapy.

[61]  F. Schluenzen,et al.  Structural basis for the interaction of antibiotics with the peptidyl transferase centre in eubacteria , 2001, Nature.

[62]  S. Krudsood,et al.  A randomized clinical trial of combinations of artesunate and azithromycin for treatment of uncomplicated Plasmodium falciparum malaria in Thailand. , 2000, The Southeast Asian journal of tropical medicine and public health.

[63]  H. Basri,et al.  Malaria prophylaxis using azithromycin: a double-blind, placebo-controlled trial in Irian Jaya, Indonesia. , 1999, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[64]  J. Berman,et al.  Successful double-blinded, randomized, placebo-controlled field trial of azithromycin and doxycycline as prophylaxis for malaria in western Kenya. , 1998, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America.

[65]  W. Yuan,et al.  mefE is necessary for the erythromycin-resistant M phenotype in Streptococcus pneumoniae , 1997, Antimicrobial agents and chemotherapy.

[66]  J. Sutcliffe,et al.  Streptococcus pneumoniae and Streptococcus pyogenes resistant to macrolides but sensitive to clindamycin: a common resistance pattern mediated by an efflux system , 1996, Antimicrobial agents and chemotherapy.

[67]  Geoffrey I. McFadden,et al.  Plastid in human parasites , 1996, Nature.

[68]  T. Schaberg,et al.  Azithromycin--review of key chemical, pharmacokinetic and microbiological features. , 1996, The Journal of antimicrobial chemotherapy.

[69]  S. Hopkins Clinical toleration and safety of azithromycin. , 1991, The American journal of medicine.

[70]  A. Cowman,et al.  Amino acid changes linked to pyrimethamine resistance in the dihydrofolate reductase-thymidylate synthase gene of Plasmodium falciparum. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[71]  Y. Zhang,et al.  Stage-dependent inhibition of chloroquine on Plasmodium falciparum in vitro. , 1986, The Journal of parasitology.

[72]  W. Trager,et al.  Human malaria parasites in continuous culture. , 1976, Science.