#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

malERA: An updated research agenda for diagnostics, drugs, vaccines, and vector control in malaria elimination and eradication


David Kaslow and colleagues examine the progress in reasearch for diagnostics, drugs, vaccines, and vector control in malaria elimination and eradication and propose a research agenda.


Vyšlo v časopise: malERA: An updated research agenda for diagnostics, drugs, vaccines, and vector control in malaria elimination and eradication. PLoS Med 14(11): e32767. doi:10.1371/journal.pmed.1002455
Kategorie: Collection Review
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pmed.1002455

Souhrn

David Kaslow and colleagues examine the progress in reasearch for diagnostics, drugs, vaccines, and vector control in malaria elimination and eradication and propose a research agenda.


Zdroje

1. World Health Organization. Eliminating malaria Geneva: WHO; 2016 [Available from: http://apps.who.int/iris/bitstream/10665/205565/1/WHO_HTM_GMP_2016.3_eng.pdf?ua=1]

2. World Health Organization. Global technical strategy for malaria 2016–2030 Geneva: WHO; 2015 [Available from: http://apps.who.int/iris/bitstream/10665/176712/1/9789241564991_eng.pdf]

3. malERA Consultative Group on Vector Control. A research agenda for malaria eradication: vector control. PLoS Med. 2011;8(1):e1000401. doi: 10.1371/journal.pmed.1000401 21311587

4. malERA Consultative Group on Vaccines. A research agenda for malaria eradication: vaccines. PLoS Med. 2011;8(1):e1000398. doi: 10.1371/journal.pmed.1000398 21311586

5. malERA Consultative Group on Drugs. A research agenda for malaria eradication: drugs. PLoS Med. 2011;8(1):e1000402. doi: 10.1371/journal.pmed.1000402 21311580

6. malERA Consultative Group on Diagnoses and Diagnostics. A research agenda for malaria eradication: diagnoses and diagnostics. PLoS Med. 2011;8(1):e1000396. doi: 10.1371/journal.pmed.1000396 21311583

7. malERA Refresh Consultative Panel on Combination Interventions and Modelling. malERA: An updated research agenda on combination interventions and modelling for malaria elimination and eradication. PLoS Med. 2017;14(11):e1002453. doi: 10.1371/journal.pmed.1002453

8. malERA Refresh Consultative Panel on Insecticide and Drug Resistance. malERA: An updated research agenda for insecticide and drug resistance in malaria elimination and eradication. PLoS Med. 2017;14(11):e1002450. doi: 10.1371/journal.pmed.1002450

9. malERA Refresh Consultative Panel on Health Systems and Policy Research. malERA: An updated research agenda for health systems, policy and implementation research in malaria elimination and eradication. PLoS Med. 2017;14(11):e1002454. doi: 10.1371/journal.pmed.1002454

10. malERA Refresh Consultative Panel on Basic Science and Enabling Technologies. malERA: An updated research agenda on basic science and enabling technologies in malaria elimination and eradication. PLoS Med. 2017;14(11):e1002451. doi: 10.1371/journal.pmed.1002451

11. Rabinovich R, Drakeley C, Djimde AA, Hall BF, Hay S, Hemingway J, et al. malERA: An updated research agenda for malaria elimination and eradication. PLoS Med. 2017;14(11):e1002456. doi: 10.1371/journal.pmed.1002456

12. World Health Organization. World malaria report Geneva: WHO; 2015 [Available from: http://apps.who.int/iris/bitstream/10665/200018/1/9789241565158_eng.pdf?ua=1]

13. Abba K, Kirkham AJ, Olliaro PL, Deeks JJ, Donegan S, Garner P, et al. Rapid diagnostic tests for diagnosing uncomplicated non-falciparum or Plasmodium vivax malaria in endemic countries. Cochrane Database Syst Rev. 2014;12:CD011431.

14. World Health Organization. Malaria rapid diagnostic test performance Geneva: WHO; 2015 [Available from: http://apps.who.int/iris/bitstream/10665/204118/1/9789241510035_eng.pdf?ua=1]

15. World Health Organization. The role of mass drug administration, mass screening and treatment, and focal screening and treatment for malaria Geneva: WHO; 2015 [Available from: http://www.who.int/malaria/publications/atoz/role-of-mda-for-malaria.pdf?ua=1]

16. World Health Organization. Policy brief on single-dose primaquine as a gametocytocide in Plasmodium falciparum malaria Geneva: WHO; 2015 [Available from: http://www.who.int/malaria/publications/atoz/who_htm_gmp_2015.1.pdf]

17. Medicines for Malaria Venture. Interactive R&D portfolio Geneva: MMV; 2016 [Available from: http://www.mmv.org/research-development/interactive-rd-portfolio]

18. Llanos-Cuentas A, Lacerda MV, Rueangweerayut R, Krudsood S, Gupta SK, Kochar SK, et al. Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria (DETECTIVE): a multicentre, double-blind, randomised, phase 2b dose-selection study. Lancet. 2014;383(9922):1049–58. doi: 10.1016/S0140-6736(13)62568-4 24360369

19. Leong FJ, Zhao R, Zeng S, Magnusson B, Diagana TT, Pertel P. A first-in-human randomized, double-blind, placebo-controlled, single- and multiple-ascending oral dose study of novel Imidazolopiperazine KAF156 to assess its safety, tolerability, and pharmacokinetics in healthy adult volunteers. Antimicrob Agents Chemother. 2014;58(11):6437–43. doi: 10.1128/AAC.03478-14 25136017

20. Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505(7481):50–5. doi: 10.1038/nature12876 24352242

21. Menard D, Khim N, Beghain J, Adegnika AA, Shafiul-Alam M, Amodu O, et al. A worldwide map of Plasmodium falciparum K13-propeller polymorphisms. N Engl J Med. 2016;374(25):2453–64. doi: 10.1056/NEJMoa1513137 27332904

22. European Medicines Agency. Assessment report: Mosquirix™ London: EMA; 2015 [Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Medicine_for_use_outside_EU/2015/10/WC500194577.pdf]

23. Agnandji ST, Fernandes JF, Bache EB, Ramharter M. Clinical development of RTS,S/AS malaria vaccine: a systematic review of clinical Phase I-III trials. Future Microbiol. 2015;10(10):1553–78. doi: 10.2217/fmb.15.90 26437872

24. Kaslow DC, Biernaux S. RTS S: Toward a first landmark on the malaria vaccine technology roadmap. Vaccine. 2015;33(52):7425–32. doi: 10.1016/j.vaccine.2015.09.061 26431982

25. RTS S, Clinical Trials Partnership,. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386(9988):31–45. doi: 10.1016/S0140-6736(15)60721-8 25913272

26. World Health Organization. Global Advisory Committee on Vaccine Safety, 10–11 June 2015 Geneva: WHO; 2015 [Available from: http://www.who.int/vaccine_safety/committee/reports/Jun_2015/en/.]

27. Penny MA, Verity R, Bever CA, Sauboin C, Galactionova K, Flasche S, et al. Public health impact and cost-effectiveness of the RTS,S/AS01 malaria vaccine: a systematic comparison of predictions from four mathematical models. Lancet. 2016;387(10016):367–75. doi: 10.1016/S0140-6736(15)00725-4 26549466

28. World Health Organization. Questions and answers on malaria vaccines Geneva: WHO; 2016 [Available from: http://www.who.int/immunization/research/development/malaria_vaccine_qa/en/.]

29. Greenwood B, Doumbo OK. Implementation of the malaria candidate vaccine RTS,S/AS01. Lancet. 2016;387(10016):318–9. doi: 10.1016/S0140-6736(15)00807-7 26549465

30. Stoute JA, Slaoui M, Heppner DG, Momin P, Kester KE, Desmons P, et al. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N Engl J Med. 1997;336(2):86–91. doi: 10.1056/NEJM199701093360202 8988885

31. Regules JA, Cicatelli SB, Bennett JW, Paolino KM, Twomey PS, Moon JE, et al. Fractional third and fourth dose of RTS,S/AS01 malaria candidate vaccine: a phase 2a controlled human malaria infection and immunogenicity study. J Infect Dis. 2016;214:762–71. doi: 10.1093/infdis/jiw237 27296848

32. Gosling R, von Seidlein L. The Future of the RTS,S/AS01 Malaria Vaccine: An Alternative Development Plan. PLoS Med. 2016;13(4):e1001994. doi: 10.1371/journal.pmed.1001994 27070151

33. White MT, Verity R, Churcher TS, Ghani AC. Vaccine approaches to malaria control and elimination: Insights from mathematical models. Vaccine. 2015;33(52):7544–50. doi: 10.1016/j.vaccine.2015.09.099 26476361

34. World Health Organization. Malaria vaccine technology roadmap: November 2013 Geneva: WHO; 2013 [Available from: http://www.who.int/immunization/topics/malaria/vaccine_roadmap/TRM_update_nov13.pdf?ua=1]

35. World Health Organization. Tables of malaria vaccine projects globally "The Rainbow Tables" Geneva: WHO; 2016 [Available from: http://www.who.int/immunization/research/development/Rainbow_tables/en/.]

36. Mueller I, Shakri AR, Chitnis CE. Development of vaccines for Plasmodium vivax malaria. Vaccine. 2015;33(52):7489–95. doi: 10.1016/j.vaccine.2015.09.060 26428453

37. International Vector Control Consortium. WHO recommends Syngenta's new long-lasting insecticide formulation Liverpool: IVCC; 2013 [Available from: http://www.ivcc.com/news-and-media/news/who-recommends-syngentas-new-long-lasting-insecticide-formulation]

38. International Vector Control Consortium. K-Othrine Polyzone Liverpool: IVCC; 2016 [Available from: http://www.ivcc.com/news-and-media/news/bayer-cropscience-and-ivcc-offer-new-tool-for-malaria-control]

39. Djenontin A, Ahoua Alou LP, Koffi A, Zogo B, Duarte E, N'Guessan R, et al. Insecticidal and sterilizing effect of Olyset Duo(R), a permethrin and pyriproxyfen mixture net against pyrethroid-susceptible and -resistant strains of Anopheles gambiae s.s.: a release-recapture assay in experimental huts. Parasite. 2015;22:27. doi: 10.1051/parasite/2015027 26489479

40. Koffi AA, Ahoua Alou LP, Djenontin A, Kabran JP, Dosso Y, Kone A, et al. Efficacy of Olyset(R) Duo, a permethrin and pyriproxyfen mixture net against wild pyrethroid-resistant Anopheles gambiae s.s. from Cote d'Ivoire: an experimental hut trial. Parasite. 2015;22:28. doi: 10.1051/parasite/2015028 26489480

41. N'Guessan R, Ngufor C, Kudom AA, Boko P, Odjo A, Malone D, et al. Mosquito nets treated with a mixture of chlorfenapyr and alphacypermethrin control pyrethroid resistant Anopheles gambiae and Culex quinquefasciatus mosquitoes in West Africa. PLoS ONE. 2014;9(2):e87710. doi: 10.1371/journal.pone.0087710 24498360

42. Innovation to Impact. [Available from: http://innovationtoimpact.org/.]

43. World Health Organization. Innovation to Impact–WHO change plan for strengthening innovation, quality and use of vector-control tools Geneva: WHO; 2015 [Available from: http://www.who.int/malaria/mpac/mpac-sept2015-vector-control-innovation.pdf]

44. International Vector Control Consortium. [Available from: http://www.ivcc.com/.]

45. World Health Organization. Larval source management—a supplementary measure for malaria vector control: an operational manual Geneva: WHO; 2013 [Available from: http://apps.who.int/iris/bitstream/10665/85379/1/9789241505604_eng.pdf?ua=1]

46. Bousema T, Dinglasan RR, Morlais I, Gouagna LC, van Warmerdam T, Awono-Ambene PH, et al. Mosquito feeding assays to determine the infectiousness of naturally infected Plasmodium falciparum gametocyte carriers. PLoS ONE. 2012;7(8):e42821. doi: 10.1371/journal.pone.0042821 22936993

47. Churcher TS, Bousema T, Walker M, Drakeley C, Schneider P, Ouedraogo AL, et al. Predicting mosquito infection from Plasmodium falciparum gametocyte density and estimating the reservoir of infection. Elife. 2013;2:e00626. doi: 10.7554/eLife.00626 23705071

48. Ouédraogo AL, Guelbéogo WM, Cohuet A, Morlais I, King JG, Gonçalves BP, et al. A protocol for membrane feeding assays to determine the infectiousness of P. falciparum naturally infected individuals to Anopheles gambiae. MWJ. 2013;4(16):1–4.

49. Li T, Eappen AG, Richman AM, Billingsley PF, Abebe Y, Li M, et al. Robust, reproducible, industrialized, standard membrane feeding assay for assessing the transmission blocking activity of vaccines and drugs against Plasmodium falciparum. Malar J. 2015;14:150. doi: 10.1186/s12936-015-0665-8 25890243

50. Laurens MB, Duncan CJ, Epstein JE, Hill AV, Komisar JL, Lyke KE, et al. A consultation on the optimization of controlled human malaria infection by mosquito bite for evaluation of candidate malaria vaccines. Vaccine. 2012;30(36):5302–4. doi: 10.1016/j.vaccine.2012.04.088 22659449

51. Spring M, Polhemus M, Ockenhouse C. Controlled human malaria infection. J Infect Dis. 2014;209 Suppl 2:S40–5.

52. Gomez-Perez GP, Legarda A, Munoz J, Sim BK, Ballester MR, Dobano C, et al. Controlled human malaria infection by intramuscular and direct venous inoculation of cryopreserved Plasmodium falciparum sporozoites in malaria-naive volunteers: effect of injection volume and dose on infectivity rates. Malar J. 2015;14:306. doi: 10.1186/s12936-015-0817-x 26245196

53. Mordmuller B, Supan C, Sim KL, Gomez-Perez GP, Ospina Salazar CL, Held J, et al. Direct venous inoculation of Plasmodium falciparum sporozoites for controlled human malaria infection: a dose-finding trial in two centres. Malar J. 2015;14:117. doi: 10.1186/s12936-015-0628-0 25889522

54. Engwerda CR, Minigo G, Amante FH, McCarthy JS. Experimentally induced blood stage malaria infection as a tool for clinical research. Trends Parasitol. 2012;28(11):515–21. doi: 10.1016/j.pt.2012.09.001 23041118

55. Krause A, Dingemanse J, Mathis A, Marquart L, Mohrle JJ, McCarthy JS. Pharmacokinetic/pharmacodynamic modelling of the antimalarial effect of Actelion-451840 in an induced blood stage malaria study in healthy subjects. Br J Clin Pharmacol. 2016;82:412–21. doi: 10.1111/bcp.12962 27062080

56. Malaria Vaccine Initiative. Transmission-blocking vaccines (TBVs) against malaria Seattle: MVI; 2016 [Available from: http://www.malariavaccine.org/sites/www.malariavaccine.org/files/content/resource/files/MVI-TBVfactsheetFINAL_20160420.pdf]

57. McCarthy JS, Marquart L, Sekuloski S, Trenholme K, Elliott S, Griffin P, et al. Linking murine and human Plasmodium falciparum challenge models in a translational path for antimalarial drug development. Antimicrob Agents Chemother. 2016;60:3669–75. doi: 10.1128/AAC.02883-15 27044554

58. Siu E, Ploss A. Modeling malaria in humanized mice: opportunities and challenges. Ann N Y Acad Sci. 2015;1342:29–36. doi: 10.1111/nyas.12618 25678404

59. Miguel-Blanco C, Lelievre J, Delves MJ, Bardera AI, Presa JL, Lopez-Barragan MJ, et al. Imaging-based high-throughput screening assay to identify new molecules with transmission-blocking potential against Plasmodium falciparum female gamete formation. Antimicrob Agents Chemother. 2015;59(6):3298–305. doi: 10.1128/AAC.04684-14 25801574

60. Asare EO, Tompkins AM, Bomblies A. A regional model for malaria vector developmental habitats evaluated using explicit, pond-resolving surface hydrology simulations. PLoS ONE. 2016;11(3):e0150626. doi: 10.1371/journal.pone.0150626 27003834

61. Chitnis N, Schapira A, Smith T, Steketee R. Comparing the effectiveness of malaria vector-control interventions through a mathematical model. Am J Trop Med Hyg. 2010;83(2):230–40. doi: 10.4269/ajtmh.2010.09-0179 20682861

62. Ngwa GA, Wankah TT, Fomboh-Nforba MY, Ngonghala CN, Teboh-Ewungkem MI. On a reproductive stage-structured model for the population dynamics of the malaria vector. Bull Math Biol. 2014;76(10):2476–516. doi: 10.1007/s11538-014-0021-0 25234336

63. World Health Organization. Policy brief on malaria diagnostics in low-transmission settings Geneva: WHO; 2014 [Available from: http://www.who.int/malaria/publications/atoz/malaria-diagnosis-low-transmission-settings-sep2014.pdf]

64. DIAMETER (Diagnostics for Malaria Elimination Toward Eradication). Target product profile: point-of-care malaria infection detection test for rapid detection of low-density, subclinical malaria infections Seattle: PATH; 2014 [Available from: http://sites.path.org/dx/files/2012/11/DIAMETER_IDT_TPP_FINAL_forwebsite.pdf]

65. Gamboa D, Ho MF, Bendezu J, Torres K, Chiodini PL, Barnwell JW, et al. A large proportion of P. falciparum isolates in the Amazon region of Peru lack pfhrp2 and pfhrp3: implications for malaria rapid diagnostic tests. PLoS ONE. 2010;5(1):e8091. doi: 10.1371/journal.pone.0008091 20111602

66. Akinyi S, Hayden T, Gamboa D, Torres K, Bendezu J, Abdallah JF, et al. Multiple genetic origins of histidine-rich protein 2 gene deletion in Plasmodium falciparum parasites from Peru. Sci Rep. 2013;3:2797. doi: 10.1038/srep02797 24077522

67. Cheng Q, Gatton ML, Barnwell J, Chiodini P, McCarthy J, Bell D, et al. Plasmodium falciparum parasites lacking histidine-rich protein 2 and 3: a review and recommendations for accurate reporting. Malar J. 2014;13:283. doi: 10.1186/1475-2875-13-283 25052298

68. Bharti PK, Chandel HS, Ahmad A, Krishna S, Udhayakumar V, Singh N. Prevalence of pfhrp2 and/or pfhrp3 gene deletion in Plasmodium falciparum population in eight highly endemic states in india. PLoS ONE. 2016;11(8):e0157949. doi: 10.1371/journal.pone.0157949 27518538

69. Deme AB, Park DJ, Bei AK, Sarr O, Badiane AS, Gueye Pel H, et al. Analysis of pfhrp2 genetic diversity in Senegal and implications for use of rapid diagnostic tests. Malar J. 2014;13:34. doi: 10.1186/1475-2875-13-34 24472178

70. Parr JB, Belson C, Patel JC, Hoffman IF, Kamthunzi P, Martinson F, et al. Estimation of Plasmodium falciparum transmission intensity in lilongwe, malawi, by microscopy, rapid diagnostic testing, and nucleic acid detection. Am J Trop Med Hyg. 2016;95(2):373–7. doi: 10.4269/ajtmh.16-0156 27325802

71. World Health Organization. False-negative RDT results and implications of new reports of P. falciparum histidine-rich protein 2/3 gene deletions Geneva: WHO; 2016 [Available from: http://www.who.int/malaria/publications/atoz/who-htm-gmp-2016.4.pdf]

72. Kobayashi T, Gamboa D, Ndiaye D, Cui L, Sutton PL, Vinetz JM. Malaria diagnosis across the International Centers of Excellence for Malaria Research: platforms, performance, and standardization. Am J Trop Med Hyg. 2015;93(3 Suppl):99–109.

73. malERA Refresh Consultative Panel on Characterising the Reservoir and Measuring Transmission. malERA: An updated research agenda for characterising the reservoir and measuring transmission in malaria elimination and eradication. PLoS Med. 2017;14: e1002452.

74. Scherr TF, Gupta S, Wright DW, Haselton FR. Mobile phone imaging and cloud-based analysis for standardized malaria detection and reporting. Sci Rep. 2016;6:28645. doi: 10.1038/srep28645 27345590

75. Soti DO, Kinoti SN, Omar AH, Logedi J, Mwendwa TK, Hirji Z, et al. Feasibility of an innovative electronic mobile system to assist health workers to collect accurate, complete and timely data in a malaria control programme in a remote setting in Kenya. Malar J. 2015;14:430. doi: 10.1186/s12936-015-0965-z 26530237

76. Falzon D, Timimi H, Kurosinski P, Migliori GB, Van Gemert W, Denkinger C, et al. Digital health for the End TB Strategy: developing priority products and making them work. Eur Respir J. 2016;48(1):29–45. doi: 10.1183/13993003.00424-2016 27230443

77. Ibrahim F, Thio TH, Faisal T, Neuman M. The application of biomedical engineering techniques to the diagnosis and management of tropical diseases: a review. Sensors (Basel). 2015;15(3):6947–95.

78. Das DK, Mukherjee R, Chakraborty C. Computational microscopic imaging for malaria parasite detection: a systematic review. J Microsc. 2015;260(1):1–19. doi: 10.1111/jmi.12270 26047029

79. Tay A, Pavesi A, Yazdi SR, Lim CT, Warkiani ME. Advances in microfluidics in combating infectious diseases. Biotechnol Adv. 2016;34(4):404–21. doi: 10.1016/j.biotechadv.2016.02.002 26854743

80. Taylor BJ, Howell A, Martin KA, Manage DP, Gordy W, Campbell SD, et al. A lab-on-chip for malaria diagnosis and surveillance. Malar J. 2014;13:179. doi: 10.1186/1475-2875-13-179 24885206

81. Kasetsirikul S, Buranapong J, Srituravanich W, Kaewthamasorn M, Pimpin A. The development of malaria diagnostic techniques: a review of the approaches with focus on dielectrophoretic and magnetophoretic methods. Malar J. 2016;15(1):358. doi: 10.1186/s12936-016-1400-9 27405995

82. Britton S, Cheng Q, McCarthy JS. Novel molecular diagnostic tools for malaria elimination: a review of options from the point of view of high-throughput and applicability in resource limited settings. Malar J. 2016;15:88. doi: 10.1186/s12936-016-1158-0 26879936

83. Patel JC, Lucchi NW, Srivastava P, Lin JT, Sug-Aram R, Aruncharus S, et al. Field evaluation of a real-time fluorescence loop-mediated isothermal amplification assay, RealAmp, for the diagnosis of malaria in Thailand and India. J Infect Dis. 2014;210(8):1180–7. doi: 10.1093/infdis/jiu252 24795480

84. Cook J, Aydin-Schmidt B, Gonzalez IJ, Bell D, Edlund E, Nassor MH, et al. Loop-mediated isothermal amplification (LAMP) for point-of-care detection of asymptomatic low-density malaria parasite carriers in Zanzibar. Malar J. 2015;14:43. doi: 10.1186/s12936-015-0573-y 25627037

85. Sema M, Alemu A, Bayih AG, Getie S, Getnet G, Guelig D, et al. Evaluation of non-instrumented nucleic acid amplification by loop-mediated isothermal amplification (NINA-LAMP) for the diagnosis of malaria in Northwest Ethiopia. Malar J. 2015;14:44. doi: 10.1186/s12936-015-0559-9 25626339

86. Waggoner JJ, Abeynayake J, Balassiano I, Lefterova M, Sahoo MK, Liu Y, et al. Multiplex nucleic acid amplification test for diagnosis of dengue fever, malaria, and leptospirosis. J Clin Microbiol. 2014;52(6):2011–8. doi: 10.1128/JCM.00341-14 24671788

87. Chua KH, Lee PC, Chai HC. Development of insulated isothermal PCR for rapid on-site malaria detection. Malar J. 2016;15(1):134.

88. Foundation for Innovative New Diagnostics. Acute febrile syndrome strategy Geneva: FIND; 2012 [Available from: http://r4d.dfid.gov.uk/PDF/Outputs/FIND/0031-FIND-NMFI-document-print-inhouse.pdf]

89. Singh R, Singh DP, Gupta R, Savargaonkar D, Singh OP, Nanda N, et al. Comparison of three PCR-based assays for the non-invasive diagnosis of malaria: detection of Plasmodium parasites in blood and saliva. Eur J Clin Microbiol Infect Dis. 2014;33(9):1631–9. doi: 10.1007/s10096-014-2121-z 24792127

90. Oguonu T, Shu E, Ezeonwu BU, Lige B, Derrick A, Umeh RE, et al. The performance evaluation of a urine malaria test (UMT) kit for the diagnosis of malaria in individuals with fever in south-east Nigeria: cross-sectional analytical study. Malar J. 2014;13:403. doi: 10.1186/1475-2875-13-403 25316216

91. Berna AZ, McCarthy JS, Wang RX, Saliba KJ, Bravo FG, Cassells J, et al. Analysis of breath specimens for biomarkers of Plasmodium falciparum infection. J Infect Dis. 2015;212(7):1120–8. doi: 10.1093/infdis/jiv176 25810441

92. Lukianova-Hleb E, Bezek S, Szigeti R, Khodarev A, Kelley T, Hurrell A, et al. Transdermal diagnosis of malaria using vapor nanobubbles. Emerg Infect Dis. 2015;21(7):1122–7. doi: 10.3201/eid2107.150089 26079141

93. Djimde AA, Maiga AW, Ouologuem D, Fofana B, Sagara I, Dembele D, et al. Gametocyte clearance dynamics following oral artesunate treatment of uncomplicated falciparum malaria in Malian children. Parasite. 2016;23:3. doi: 10.1051/parasite/2016003 26839003

94. Mwingira F, Genton B, Kabanywanyi AN, Felger I. Comparison of detection methods to estimate asexual Plasmodium falciparum parasite prevalence and gametocyte carriage in a community survey in Tanzania. Malar J. 2014;13:433. doi: 10.1186/1475-2875-13-433 25404207

95. Wampfler R, Mwingira F, Javati S, Robinson L, Betuela I, Siba P, et al. Strategies for detection of Plasmodium species gametocytes. PLoS ONE. 2013;8(9):e76316. doi: 10.1371/journal.pone.0076316 24312682

96. Ouedraogo AL, Schneider P, de Kruijf M, Nebie I, Verhave JP, Cuzin-Ouattara N, et al. Age-dependent distribution of Plasmodium falciparum gametocytes quantified by Pfs25 real-time QT-NASBA in a cross-sectional study in Burkina Faso. Am J Trop Med Hyg. 2007;76(4):626–30. 17426160

97. Schneider P, Reece SE, van Schaijk BC, Bousema T, Lanke KH, Meaden CS, et al. Quantification of female and male Plasmodium falciparum gametocytes by reverse transcriptase quantitative PCR. Mol Biochem Parasitol. 2015;199(1–2):29–33. doi: 10.1016/j.molbiopara.2015.03.006 25827756

98. Hisaeda H, Stowers AW, Tsuboi T, Collins WE, Sattabongkot JS, Suwanabun N, et al. Antibodies to malaria vaccine candidates Pvs25 and Pvs28 completely block the ability of Plasmodium vivax to infect mosquitoes. Infect Immun. 2000;68(12):6618–23. 11083773

99. World Health Organization. Emergency response to artemisinin resistance in the Greater Mekong subregion. Regional framework for action 2013–2015 Geneva: WHO; 2013 [Available from: http://www.who.int/malaria/publications/atoz/9789241505321/en/]

100. Rao PN, Uplekar S, Kayal S, Mallick PK, Bandyopadhyay N, Kale S, et al. A method for amplicon deep sequencing of drug resistance genes in Plasmodium falciparum clinical isolates from India. J Clin Microbiol. 2016;54(6):1500–11. doi: 10.1128/JCM.00235-16 27008882

101. Nankoberanyi S, Mbogo GW, LeClair NP, Conrad MD, Tumwebaze P, Tukwasibwe S, et al. Validation of the ligase detection reaction fluorescent microsphere assay for the detection of Plasmodium falciparum resistance mediating polymorphisms in Uganda. Malar J. 2014;13:95. doi: 10.1186/1475-2875-13-95 24629020

102. Amato R, Lim P, Miotto O, Amaratunga C, Dek D, Pearson RD, et al. Genetic markers associated with dihydroartemisinin-piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis. 2017;17:164–73. doi: 10.1016/S1473-3099(16)30409-1 27818095

103. Witkowski B, Duru V, Khim N, Ross LS, Saintpierre B, Beghain J, et al. A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: a phenotype-genotype association study. Lancet Infect Dis. 2017;17:174–83. doi: 10.1016/S1473-3099(16)30415-7 27818097

104. White MT, Karl S, Battle KE, Hay SI, Mueller I, Ghani AC. Modelling the contribution of the hypnozoite reservoir to Plasmodium vivax transmission. Elife. 2014;3:e04692.

105. Ley B, Luter N, Espino FE, Devine A, Kalnoky M, Lubell Y, et al. The challenges of introducing routine G6PD testing into radical cure: a workshop report. Malar J. 2015;14:377. doi: 10.1186/s12936-015-0896-8 26416229

106. World Health Organization. Guidelines for the treatment of malaria, third edition Geneva: WHO; 2015 [Available from: http://apps.who.int/iris/bitstream/10665/162441/1/9789241549127_eng.pdf]

107. Graves PM, Gelband H, Garner P. Primaquine or other 8-aminoquinoline for reducing Plasmodium falciparum transmission. Cochrane Database Syst Rev. 2015(2):CD008152. doi: 10.1002/14651858.CD008152.pub4 25693791

108. Salinas JL, Kissinger JC, Jones DP, Galinski MR. Metabolomics in the fight against malaria. Mem Inst Oswaldo Cruz. 2014;109(5):589–97. doi: 10.1590/0074-0276140043 25185001

109. Chavchich M, Van Breda K, Rowcliffe K, Diagana TT, Edstein MD. The spiroindolone KAE609 does not induce dormant ring stages in Plasmodium falciparum parasites. Antimicrob Agents Chemother. 2016;60(9):5167–74. doi: 10.1128/AAC.02838-15 27297484

110. Burrows JN, Duparc S, Gutteridge WE, Hooft van Huijsduijnen R, Kaszubska W, Macintyre F, et al. New developments in anti-malarial target candidate and product profiles. Malar J. 2017;16(1):26. doi: 10.1186/s12936-016-1675-x 28086874

111. Artzy-Randrup Y, Alonso D, Pascual M. Transmission intensity and drug resistance in malaria population dynamics: implications for climate change. PLoS ONE. 2010;5(10):e13588. doi: 10.1371/journal.pone.0013588 21060886

112. Hastings IM, Watkins WM. Intensity of malaria transmission and the evolution of drug resistance. Acta Trop. 2005;94(3):218–29. doi: 10.1016/j.actatropica.2005.04.003 15847846

113. Dondorp AM, Fanello CI, Hendriksen IC, Gomes E, Seni A, Chhaganlal KD, et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010;376(9753):1647–57. doi: 10.1016/S0140-6736(10)61924-1 21062666

114. Dondorp A, Nosten F, Stepniewska K, Day N, White N, South East Asian Quinine Artesunate Malaria Trial g. Artesunate versus quinine for treatment of severe falciparum malaria: a randomised trial. Lancet. 2005;366(9487):717–25. doi: 10.1016/S0140-6736(05)67176-0 16125588

115. Jeeyapant A, Kingston HW, Plewes K, Maude RJ, Hanson J, Herdman MT, et al. Defining surrogate endpoints for clinical trials in severe falciparum malaria. PLoS ONE. 2017;12(1):e0169307. doi: 10.1371/journal.pone.0169307 28052109

116. Cheah PY, Parker M, Dondorp AM. Development of drugs for severe malaria in children. Int Health. 2016;8(5):313–6. doi: 10.1093/inthealth/ihw038 27620923

117. Abdul-Ghani R, Beier JC. Strategic use of antimalarial drugs that block falciparum malaria parasite transmission to mosquitoes to achieve local malaria elimination. Parasitol Res. 2014;113(10):3535–46. doi: 10.1007/s00436-014-4091-6 25185662

118. Dicko A, Brown JM, Diawara H, Baber I, Mahamar A, Soumare HM, et al. Primaquine to reduce transmission of Plasmodium falciparum malaria in Mali: a single-blind, dose-ranging, adaptive randomised phase 2 trial. Lancet Infect Dis. 2016;16:674–84. doi: 10.1016/S1473-3099(15)00479-X 26906747

119. Goncalves BP, Tiono AB, Ouedraogo A, Guelbeogo WM, Bradley J, Nebie I, et al. Single low dose primaquine to reduce gametocyte carriage and Plasmodium falciparum transmission after artemether-lumefantrine in children with asymptomatic infection: a randomised, double-blind, placebo-controlled trial. BMC Med. 2016;14:40. doi: 10.1186/s12916-016-0581-y 26952094

120. Bolscher JM, Koolen KM, van Gemert GJ, van de Vegte-Bolmer MG, Bousema T, Leroy D, et al. A combination of new screening assays for prioritization of transmission-blocking antimalarials reveals distinct dynamics of marketed and experimental drugs. J Antimicrob Chemother. 2015;70(5):1357–66. doi: 10.1093/jac/dkv003 25667405

121. Plouffe DM, Wree M, Du AY, Meister S, Li F, Patra K, et al. High-throughput assay and discovery of small molecules that interrupt malaria transmission. Cell Host Microbe. 2016;19(1):114–26. doi: 10.1016/j.chom.2015.12.001 26749441

122. Lucantoni L, Fidock DA, Avery VM. Luciferase-based, high-throughput assay for screening and profiling transmission-blocking compounds against Plasmodium falciparum gametocytes. Antimicrob Agents Chemother. 2016;60(4):2097–107. doi: 10.1128/AAC.01949-15 26787698

123. Almela MJ, Lozano S, Lelievre J, Colmenarejo G, Coteron JM, Rodrigues J, et al. A new set of chemical starting points with Plasmodium falciparum transmission-blocking potential for antimalarial drug discovery. PLoS ONE. 2015;10(8):e0135139. doi: 10.1371/journal.pone.0135139 26317851

124. Lelievre J, Almela MJ, Lozano S, Miguel C, Franco V, Leroy D, et al. Activity of clinically relevant antimalarial drugs on Plasmodium falciparum mature gametocytes in an ATP bioluminescence "transmission blocking" assay. PLoS ONE. 2012;7(4):e35019. doi: 10.1371/journal.pone.0035019 22514702

125. Roncales M, Vidal-Mas J, Leroy D, Herreros E. Comparison and optimization of different methods for the in vitro production of Plasmodium falciparum gametocytes. J Parasitol Res. 2012;2012:927148. doi: 10.1155/2012/927148 22523643

126. Burrows JN, van Huijsduijnen RH, Mohrle JJ, Oeuvray C, Wells TN. Designing the next generation of medicines for malaria control and eradication. Malar J. 2013;12:187. doi: 10.1186/1475-2875-12-187 23742293

127. Phillips MA, Lotharius J, Marsh K, White J, Dayan A, White KL, et al. A long-duration dihydroorotate dehydrogenase inhibitor (DSM265) for prevention and treatment of malaria. Sci Transl Med. 2015;7(296):296ra111. doi: 10.1126/scitranslmed.aaa6645 26180101

128. Robinson LJ, Wampfler R, Betuela I, Karl S, White MT, Li Wai Suen CS, et al. Strategies for understanding and reducing the Plasmodium vivax and Plasmodium ovale hypnozoite reservoir in Papua New Guinean children: a randomised placebo-controlled trial and mathematical model. PLoS Med. 2015;12(10):e1001891. doi: 10.1371/journal.pmed.1001891 26505753

129. White NJ. Why do some primate malarias relapse? Trends Parasitol. 2016;32(12):918–20. doi: 10.1016/j.pt.2016.08.014 27743866

130. John GK, Douglas NM, von Seidlein L, Nosten F, Baird JK, White NJ, et al. Primaquine radical cure of Plasmodium vivax: a critical review of the literature. Malar J. 2012;11:280. doi: 10.1186/1475-2875-11-280 22900786

131. Wells TN, Burrows JN, Baird JK. Targeting the hypnozoite reservoir of Plasmodium vivax: the hidden obstacle to malaria elimination. Trends Parasitol. 2010;26(3):145–51. doi: 10.1016/j.pt.2009.12.005 20133198

132. March S, Ng S, Velmurugan S, Galstian A, Shan J, Logan DJ, et al. A microscale human liver platform that supports the hepatic stages of Plasmodium falciparum and vivax. Cell Host Microbe. 2013;14(1):104–15. doi: 10.1016/j.chom.2013.06.005 23870318

133. Vaughan AM, Kappe SH, Ploss A, Mikolajczak SA. Development of humanized mouse models to study human malaria parasite infection. Future Microbiol. 2012;7(5):657–65. doi: 10.2217/fmb.12.27 22568719

134. Cairns M, Roca-Feltrer A, Garske T, Wilson AL, Diallo D, Milligan PJ, et al. Estimating the potential public health impact of seasonal malaria chemoprevention in African children. Nat Commun. 2012;3:881. doi: 10.1038/ncomms1879 22673908

135. Zongo I, Milligan P, Compaore YD, Some AF, Greenwood B, Tarning J, et al. Randomized noninferiority trial of dihydroartemisinin-piperaquine compared with sulfadoxine-pyrimethamine plus amodiaquine for seasonal malaria chemoprevention in Burkina Faso. Antimicrob Agents Chemother. 2015;59(8):4387–96. doi: 10.1128/AAC.04923-14 25918149

136. Griffin JT, Bhatt S, Sinka ME, Gething PW, Lynch M, Patouillard E, et al. Potential for reduction of burden and local elimination of malaria by reducing Plasmodium falciparum malaria transmission: a mathematical modelling study. Lancet Infect Dis. 2016;16(4):465–72. doi: 10.1016/S1473-3099(15)00423-5 26809816

137. Chaccour CJ, Rabinovich NR, Slater H, Canavati SE, Bousema T, Lacerda M, et al. Establishment of the Ivermectin Research for Malaria Elimination Network: updating the research agenda. Malar J. 2015;14:243. doi: 10.1186/s12936-015-0691-6 26068560

138. Slater HC, Walker PG, Bousema T, Okell LC, Ghani AC. The potential impact of adding ivermectin to a mass treatment intervention to reduce malaria transmission: a modelling study. J Infect Dis. 2014;210(12):1972–80. doi: 10.1093/infdis/jiu351 24951826

139. Chaccour CJ, Kobylinski KC, Bassat Q, Bousema T, Drakeley C, Alonso P, et al. Ivermectin to reduce malaria transmission: a research agenda for a promising new tool for elimination. Malar J. 2013;12:153. doi: 10.1186/1475-2875-12-153 23647969

140. Bogers WM, Oostermeijer H, Mooij P, Koopman G, Verschoor EJ, Davis D, et al. Potent immune responses in rhesus macaques induced by nonviral delivery of a self-amplifying RNA vaccine expressing HIV type 1 envelope with a cationic nanoemulsion. J Infect Dis. 2015;211(6):947–55. doi: 10.1093/infdis/jiu522 25234719

141. Kotta S, Khan AW, Ansari SH, Sharma RK, Ali J. Anti HIV nanoemulsion formulation: optimization and in vitro-in vivo evaluation. Int J Pharm. 2014;462(1–2):129–34. doi: 10.1016/j.ijpharm.2013.12.038 24374067

142. Jain A, Thakur K, Kush P, Jain UK. Docetaxel loaded chitosan nanoparticles: formulation, characterization and cytotoxicity studies. Int J Biol Macromol. 2014;69:546–53. doi: 10.1016/j.ijbiomac.2014.06.029 24971551

143. Mishra P, SS R, Jerobin J, Thomas J, Mukherjee A, Chandrasekaran N. Study on antimicrobial potential of neem oil nanoemulsion against Pseudomonas aeruginosa infection in Labeo rohita. Biotechnol Appl Biochem. 2014;61(5):611–9. doi: 10.1002/bab.1213 24502533

144. Campo B, Vandal O, Wesche DL, Burrows JN. Killing the hypnozoite—drug discovery approaches to prevent relapse in Plasmodium vivax. Pathog Glob Health. 2015;109(3):107–22. doi: 10.1179/2047773215Y.0000000013 25891812

145. Mikolajczak SA, Vaughan AM, Kangwanrangsan N, Roobsoong W, Fishbaugher M, Yimamnuaychok N, et al. Plasmodium vivax liver stage development and hypnozoite persistence in human liver-chimeric mice. Cell Host Microbe. 2015;17(4):526–35. doi: 10.1016/j.chom.2015.02.011 25800544

146. Imwong M, Snounou G, Pukrittayakamee S, Tanomsing N, Kim JR, Nandy A, et al. Relapses of Plasmodium vivax infection usually result from activation of heterologous hypnozoites. J Infect Dis. 2007;195(7):927–33. doi: 10.1086/512241 17330781

147. Chen N, Auliff A, Rieckmann K, Gatton M, Cheng Q. Relapses of Plasmodium vivax infection result from clonal hypnozoites activated at predetermined intervals. J Infect Dis. 2007;195(7):934–41. doi: 10.1086/512242 17330782

148. Beck HP, Wampfler R, Carter N, Koh G, Osorio L, Rueangweerayut R, et al. Estimation of the antirelapse efficacy of tafenoquine, using Plasmodium vivax genotyping. J Infect Dis. 2016;213(5):794–9. doi: 10.1093/infdis/jiv508 26500351

149. Richie TL, Billingsley PF, Sim BK, James ER, Chakravarty S, Epstein JE, et al. Progress with Plasmodium falciparum sporozoite (PfSPZ)-based malaria vaccines. Vaccine. 2015;33(52):7452–61. doi: 10.1016/j.vaccine.2015.09.096 26469720

150. Miura K. Progress and prospects for blood-stage malaria vaccines. Expert Rev Vaccines. 2016;15(6):765–81. doi: 10.1586/14760584.2016.1141680 26760062

151. Draper SJ, Angov E, Horii T, Miller LH, Srinivasan P, Theisen M, et al. Recent advances in recombinant protein-based malaria vaccines. Vaccine. 2015;33(52):7433–43. doi: 10.1016/j.vaccine.2015.09.093 26458807

152. World Health Organization. WHO Product Development for Vaccines Advisory Committee (PD-VAC) meeting– 2015 Geneva: WHO; 2015 [Available from: http://www.who.int/immunization/research/meetings_workshops/pdvac/en/.]

153. Nunes JK, Woods C, Carter T, Raphael T, Morin MJ, Diallo D, et al. Development of a transmission-blocking malaria vaccine: progress, challenges, and the path forward. Vaccine. 2014;32(43):5531–9. doi: 10.1016/j.vaccine.2014.07.030 25077422

154. Delrieu I, Leboulleux D, Ivinson K, Gessner BD, Malaria Transmission Blocking Vaccine Technical Consultation G. Design of a Phase III cluster randomized trial to assess the efficacy and safety of a malaria transmission blocking vaccine. Vaccine. 2015;33(13):1518–26. doi: 10.1016/j.vaccine.2015.01.050 25681064

155. Shimp RL Jr., Rowe C, Reiter K, Chen B, Nguyen V, Aebig J, et al. Development of a Pfs25-EPA malaria transmission blocking vaccine as a chemically conjugated nanoparticle. Vaccine. 2013;31(28):2954–62. doi: 10.1016/j.vaccine.2013.04.034 23623858

156. Li Y, Leneghan DB, Miura K, Nikolaeva D, Brian IJ, Dicks MD, et al. Enhancing immunogenicity and transmission-blocking activity of malaria vaccines by fusing Pfs25 to IMX313 multimerization technology. Sci Rep. 2016;6:18848. doi: 10.1038/srep18848 26743316

157. Bennett JW, Yadava A, Tosh D, Sattabongkot J, Komisar J, Ware LA, et al. Phase 1/2a Trial of Plasmodium vivax malaria vaccine candidate VMP001/AS01B in malaria-naive adults: safety, immunogenicity, and efficacy. PLoS Negl Trop Dis. 2016;10(2):e0004423. doi: 10.1371/journal.pntd.0004423 26919472

158. Herrera S, Fernandez OL, Vera O, Cardenas W, Ramirez O, Palacios R, et al. Phase I safety and immunogenicity trial of Plasmodium vivax CS derived long synthetic peptides adjuvanted with montanide ISA 720 or montanide ISA 51. Am J Trop Med Hyg. 2011;84(2 Suppl):12–20.

159. Bauza K, Malinauskas T, Pfander C, Anar B, Jones EY, Billker O, et al. Efficacy of a Plasmodium vivax malaria vaccine using ChAd63 and modified vaccinia Ankara expressing thrombospondin-related anonymous protein as assessed with transgenic Plasmodium berghei parasites. Infect Immun. 2014;82(3):1277–86. doi: 10.1128/IAI.01187-13 24379295

160. de Cassan SC, Shakri AR, Llewellyn D, Elias SC, Cho JS, Goodman AL, et al. Preclinical assessment of viral vectored and protein vaccines targeting the duffy-binding protein region ii of Plasmodium vivax. Front Immunol. 2015;6:348. doi: 10.3389/fimmu.2015.00348 26217340

161. McCarthy JS, Griffin PM, Sekuloski S, Bright AT, Rockett R, Looke D, et al. Experimentally induced blood-stage Plasmodium vivax infection in healthy volunteers. J Infect Dis. 2013;208(10):1688–94. doi: 10.1093/infdis/jit394 23908484

162. Rutledge G, Boehme U, Sanders M, Reid A, Maiga-Ascofare O, Djimde A, et al. Elusive Plasmodium species complete the human malaria genome set bioRxiv: Cold Spring Harbor Laboratory; 2016 [Available from: http://biorxiv.org/content/early/2016/05/12/052696.full.pdf+html]

163. Lee S, Nguyen MT. Recent advances of vaccine adjuvants for infectious diseases. Immune Netw. 2015;15(2):51–7. doi: 10.4110/in.2015.15.2.51 25922593

164. Kelley B. Industrialization of mAb production technology: the bioprocessing industry at a crossroads. MAbs. 2009;1(5):443–52. 20065641

165. Centre for Biosecurity of UPMC. Next generation monoclonal antibodies: challenges and opportunities Baltimore, MD: Centre for Biosecurity of UPMC; 2013 [Available from: http://www.upmchealthsecurity.org/our-work/pubs_archive/pubs-pdfs/2013/2013-02-04-next-gen-monoclonal-antibodies.pdf]

166. Robbie GJ, Criste R, Dall'acqua WF, Jensen K, Patel NK, Losonsky GA, et al. A novel investigational Fc-modified humanized monoclonal antibody, motavizumab-YTE, has an extended half-life in healthy adults. Antimicrob Agents Chemother. 2013;57(12):6147–53. doi: 10.1128/AAC.01285-13 24080653

167. Rasmussen SK, Naested H, Muller C, Tolstrup AB, Frandsen TP. Recombinant antibody mixtures: production strategies and cost considerations. Arch Biochem Biophys. 2012;526(2):139–45. doi: 10.1016/j.abb.2012.07.001 22820097

168. Brock PM, Fornace KM, Parmiter M, Cox J, Drakeley CJ, Ferguson HM, et al. Plasmodium knowlesi transmission: integrating quantitative approaches from epidemiology and ecology to understand malaria as a zoonosis. Parasitology. 2016;143(4):389–400. doi: 10.1017/S0031182015001821 26817785

169. Li S, Plebanski M, Smooker P, Gowans EJ. Editorial: Why vaccines to HIV, HCV, and malaria have so far failed-challenges to developing vaccines against immunoregulating pathogens. Front Microbiol. 2015;6:1318. doi: 10.3389/fmicb.2015.01318 26640461

170. Neafsey DE, Juraska M, Bedford T, Benkeser D, Valim C, Griggs A, et al. Genetic diversity and protective efficacy of the RTS,S/AS01 malaria vaccine. N Engl J Med. 2015;373(21):2025–37. doi: 10.1056/NEJMoa1505819 26488565

171. Plowe CV. Vaccine-Resistant Malaria. N Engl J Med. 2015;373(21):2082–3. doi: 10.1056/NEJMe1511955 26488465

172. Takala SL, Plowe CV. Genetic diversity and malaria vaccine design, testing and efficacy: preventing and overcoming 'vaccine resistant malaria'. Parasite Immunol. 2009;31(9):560–73. doi: 10.1111/j.1365-3024.2009.01138.x 19691559

173. Olotu A, Fegan G, Wambua J, Nyangweso G, Leach A, Lievens M, et al. Seven-year efficacy of RTS,S/AS01 malaria vaccine among young African children. N Engl J Med. 2016;374(26):2519–29. doi: 10.1056/NEJMoa1515257 27355532

174. Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526(7572):207–11. doi: 10.1038/nature15535 26375008

175. World Health Organization. Insecticide resistance Geneva: WHO; 2015 [Available from: http://www.who.int/malaria/areas/vector_control/insecticide_resistance/en/.]

176. Hemingway J, Ranson H, Magill A, Kolaczinski J, Fornadel C, Gimnig J, et al. Averting a malaria disaster: will insecticide resistance derail malaria control? Lancet. 2016;387(10029):1785–8. doi: 10.1016/S0140-6736(15)00417-1 26880124

177. World Health Organization Pesticide Evaluation Scheme. WHO recommended long-lasting insecticidal nets 2016 [Available from: http://www.who.int/whopes/Long-lasting_insecticidal_nets_April_2016.pdf?ua=1]

178. World Health Organization Pesticide Evaluation Scheme. Pesticide products under WHOPES laboratory and or field testing and evaluation Geneva: WHOPES; 2016 [Available from: http://www.who.int/whopes/Products_Under_WHOPES_Evaluation_March_2016.pdf?ua=1]

179. N'Guessan R, Asidi A, Boko P, Odjo A, Akogbeto M, Pigeon O, et al. An experimental hut evaluation of PermaNet((R)) 3.0, a deltamethrin-piperonyl butoxide combination net, against pyrethroid-resistant Anopheles gambiae and Culex quinquefasciatus mosquitoes in southern Benin. Trans R Soc Trop Med Hyg. 2010;104(12):758–65. doi: 10.1016/j.trstmh.2010.08.008 20956008

180. Pennetier C, Bouraima A, Chandre F, Piameu M, Etang J, Rossignol M, et al. Efficacy of Olyset(R) Plus, a new long-lasting insecticidal net incorporating permethrin and piperonyl-butoxide against multi-resistant malaria vectors [corrected]. PLoS ONE. 2013;8(10):e75134. doi: 10.1371/journal.pone.0075134 24116029

181. Tungu P, Magesa S, Maxwell C, Malima R, Masue D, Sudi W, et al. Evaluation of PermaNet 3.0 a deltamethrin-PBO combination net against Anopheles gambiae and pyrethroid resistant Culex quinquefasciatus mosquitoes: an experimental hut trial in Tanzania. Malar J. 2010;9:21. doi: 10.1186/1475-2875-9-21 20085631

182. Morel CM, Thang ND, Erhart A, Xa NX, Peeters Grietens K, Xuan Hung L, et al. Cost-effectiveness of long-lasting insecticide-treated hammocks in preventing malaria in South-central Vietnam. PLoS ONE. 2013;8(3):e58205. doi: 10.1371/journal.pone.0058205 23536790

183. NgenIRS. 2016 [Available from: http://www.ngenirs.org/.]

184. Knapp J, Macdonald M, Malone D, Hamon N, Richardson JH. Disruptive technology for vector control: the Innovative Vector Control Consortium and the US Military join forces to explore transformative insecticide application technology for mosquito control programmes. Malar J. 2015;14:371. doi: 10.1186/s12936-015-0907-9 26409879

185. Sibanda M, Focke W. Development of an insecticide impregnated polymer wall lining for malaria vector control. Malar J. 2014;13 (Suppl 1):80.

186. Kruger T, Sibanda MM, Focke WW, Bornman MS, de Jager C. Acceptability and effectiveness of a monofilament, polyethylene insecticide-treated wall lining for malaria control after six months in dwellings in Vhembe District, Limpopo Province, South Africa. Malar J. 2015;14:485. doi: 10.1186/s12936-015-1005-8 26628275

187. Andriessen R, Snetselaar J, Suer RA, Osinga AJ, Deschietere J, Lyimo IN, et al. Electrostatic coating enhances bioavailability of insecticides and breaks pyrethroid resistance in mosquitoes. Proc Natl Acad Sci U S A. 2015;112(39):12081–6. doi: 10.1073/pnas.1510801112 26324912

188. Briet OJ, Chitnis N. Effects of changing mosquito host searching behaviour on the cost effectiveness of a mass distribution of long-lasting, insecticidal nets: a modelling study. Malar J. 2013;12:215. doi: 10.1186/1475-2875-12-215 23802594

189. Gatton ML, Chitnis N, Churcher T, Donnelly MJ, Ghani AC, Godfray HC, et al. The importance of mosquito behavioural adaptations to malaria control in Africa. Evolution. 2013;67(4):1218–30. doi: 10.1111/evo.12063 23550770

190. Govella NJ, Chaki PP, Killeen GF. Entomological surveillance of behavioural resilience and resistance in residual malaria vector populations. Malar J. 2013;12:124. doi: 10.1186/1475-2875-12-124 23577656

191. Killeen GF, Chitnis N. Potential causes and consequences of behavioural resilience and resistance in malaria vector populations: a mathematical modelling analysis. Malar J. 2014;13:97. doi: 10.1186/1475-2875-13-97 24629066

192. Russell TL, Beebe NW, Cooper RD, Lobo NF, Burkot TR. Successful malaria elimination strategies require interventions that target changing vector behaviours. Malar J. 2013;12:56. doi: 10.1186/1475-2875-12-56 23388506

193. Sokhna C, Ndiath MO, Rogier C. The changes in mosquito vector behaviour and the emerging resistance to insecticides will challenge the decline of malaria. Clin Microbiol Infect. 2013;19(10):902–7. doi: 10.1111/1469-0691.12314 23910459

194. Sougoufara S, Diedhiou SM, Doucoure S, Diagne N, Sembene PM, Harry M, et al. Biting by Anopheles funestus in broad daylight after use of long-lasting insecticidal nets: a new challenge to malaria elimination. Malar J. 2014;13:125. doi: 10.1186/1475-2875-13-125 24678587

195. Diabate A, Tripet F. Targeting male mosquito mating behaviour for malaria control. Parasit Vectors. 2015;8:347. doi: 10.1186/s13071-015-0961-8 26113015

196. Donnelly B, Berrang-Ford L, Ross NA, Michel P. A systematic, realist review of zooprophylaxis for malaria control. Malar J. 2015;14:313. doi: 10.1186/s12936-015-0822-0 26264913

197. Durnez L, Coosemans M. Residual transmission of malaria: an old issue for new approaches Anopheles mosquitoes. In: Manguin S, editor. Anopheles mosquitoes, new insights into malaria vectors. Rijeka: InTech; 2013. p. 671–704.

198. Killeen GF. Characterizing, controlling and eliminating residual malaria transmission. Malar J. 2014;13:330. doi: 10.1186/1475-2875-13-330 25149656

199. Reddy MR, Overgaard HJ, Abaga S, Reddy VP, Caccone A, Kiszewski AE, et al. Outdoor host seeking behaviour of Anopheles gambiae mosquitoes following initiation of malaria vector control on Bioko Island, Equatorial Guinea. Malar J. 2011;10:184. doi: 10.1186/1475-2875-10-184 21736750

200. Russell TL, Govella NJ, Azizi S, Drakeley CJ, Kachur SP, Killeen GF. Increased proportions of outdoor feeding among residual malaria vector populations following increased use of insecticide-treated nets in rural Tanzania. Malar J. 2011;10:80. doi: 10.1186/1475-2875-10-80 21477321

201. Derua YA, Alifrangis M, Hosea KM, Meyrowitsch DW, Magesa SM, Pedersen EM, et al. Change in composition of the Anopheles gambiae complex and its possible implications for the transmission of malaria and lymphatic filariasis in north-eastern Tanzania. Malar J. 2012;11:188. doi: 10.1186/1475-2875-11-188 22681999

202. Mwangangi JM, Muturi EJ, Muriu SM, Nzovu J, Midega JT, Mbogo C. The role of Anopheles arabiensis and Anopheles coustani in indoor and outdoor malaria transmission in Taveta District, Kenya. Parasit Vectors. 2013;6:114. doi: 10.1186/1756-3305-6-114 23601146

203. Revay EE, Schlein Y, Tsabari O, Kravchenko V, Qualls W, De-Xue R, et al. Formulation of attractive toxic sugar bait (ATSB) with safe EPA-exempt substance significantly diminishes the Anopheles sergentii population in a desert oasis. Acta Trop. 2015;150:29–34. doi: 10.1016/j.actatropica.2015.06.018 26119042

204. Marshall JM, White MT, Ghani AC, Schlein Y, Muller GC, Beier JC. Quantifying the mosquito's sweet tooth: modelling the effectiveness of attractive toxic sugar baits (ATSB) for malaria vector control. Malar J. 2013;12:291. doi: 10.1186/1475-2875-12-291 23968494

205. Johnson BJ, Ritchie SA. The siren's song: Exploitation of female flight tones to passively capture male Aedes aegypti (Diptera: Culicidae). J Med Entomol. 2016;53(1):245–8. doi: 10.1093/jme/tjv165 26502754

206. Downes J. The swarming and mating flight of Diptera. Ann Rev Entomol. 1969;14:271–98.

207. World Health Organization. Guidelines for efficacy testing of spatial repellents: WHO; 2013 [Available from: http://apps.who.int/iris/bitstream/10665/78142/1/9789241505024_eng.pdf?ua=1]

208. Syafruddin D, Bangs MJ, Sidik D, Elyazar I, Asih PB, Chan K, et al. Impact of a spatial repellent on malaria incidence in two villages in Sumba, Indonesia. Am J Trop Med Hyg. 2014;91(6):1079–87. doi: 10.4269/ajtmh.13-0735 25311699

209. Hill N, Zhou HN, Wang P, Guo X, Carneiro I, Moore SJ. A household randomized, controlled trial of the efficacy of 0.03% transfluthrin coils alone and in combination with long-lasting insecticidal nets on the incidence of Plasmodium falciparum and Plasmodium vivax malaria in Western Yunnan Province, China. Malar J. 2014;13:208. doi: 10.1186/1475-2875-13-208 24885993

210. Tusting LS, Ippolito MM, Willey BA, Kleinschmidt I, Dorsey G, Gosling RD, et al. The evidence for improving housing to reduce malaria: a systematic review and meta-analysis. Malar J. 2015;14:209. doi: 10.1186/s12936-015-0724-1 26055986

211. Tusting LS, Thwing J, Sinclair D, Fillinger U, Gimnig J, Bonner KE, et al. Mosquito larval source management for controlling malaria. Cochrane Database Syst Rev. 2013;8:CD008923.

212. Committee on Gene Drive Research in Non-Human Organisms: Recommendations for Responsible Conduct. Gene drives on the horizon: advancing science, navigating uncertainty and aligning research with public values Washington DC: The National Academies Press; 2016 [Available from: http://www.nap.edu/catalog/23405/gene-drives-on-the-horizon-advancing-science-navigating-uncertainty-and]

213. Burt A. Heritable strategies for controlling insect vectors of disease. Philos Trans R Soc Lond B Biol Sci. 2014;369(1645):20130432. doi: 10.1098/rstb.2013.0432 24821918

214. Oliva CF, Vreysen MJ, Dupe S, Lees RS, Gilles JR, Gouagna LC, et al. Current status and future challenges for controlling malaria with the sterile insect technique: technical and social perspectives. Acta Trop. 2014;132 Suppl:S130–9. doi: 10.1016/j.actatropica.2013.11.019 24295892

215. Bourtzis K, Lees RS, Hendrichs J, Vreysen MJ. More than one rabbit out of the hat: Radiation, transgenic and symbiont-based approaches for sustainable management of mosquito and tsetse fly populations. Acta Trop. 2016;157:115–30. doi: 10.1016/j.actatropica.2016.01.009 26774684

216. Black WCt, Alphey L, James AA. Why RIDL is not SIT. Trends Parasitol. 2011;27(8):362–70. doi: 10.1016/j.pt.2011.04.004 21659002

217. Windbichler N, Menichelli M, Papathanos PA, Thyme SB, Li H, Ulge UY, et al. A synthetic homing endonuclease-based gene drive system in the human malaria mosquito. Nature. 2011;473(7346):212–5. doi: 10.1038/nature09937 21508956

218. Esvelt KM, Smidler AL, Catteruccia F, Church GM. Concerning RNA-guided gene drives for the alteration of wild populations. Elife. 2014:e03401. doi: 10.7554/eLife.03401 25035423

219. Hammond A, Galizi R, Kyrou K, Simoni A, Siniscalchi C, Katsanos D, et al. A CRISPR-Cas9 gene drive system targeting female reproduction in the malaria mosquito vector Anopheles gambiae. Nat Biotechnol. 2016;34(1):78–83. doi: 10.1038/nbt.3439 26641531

220. Gantz VM, Jasinskiene N, Tatarenkova O, Fazekas A, Macias VM, Bier E, et al. Highly efficient Cas9-mediated gene drive for population modification of the malaria vector mosquito Anopheles stephensi. Proc Natl Acad Sci U S A. 2015;112(49):E6736–43. doi: 10.1073/pnas.1521077112 26598698

221. Wilke AB, Marrelli MT. Paratransgenesis: a promising new strategy for mosquito vector control. Parasit Vectors. 2015;8:342. doi: 10.1186/s13071-015-0959-2 26104575

222. Wang S, Ghosh AK, Bongio N, Stebbings KA, Lampe DJ, Jacobs-Lorena M. Fighting malaria with engineered symbiotic bacteria from vector mosquitoes. Proc Natl Acad Sci U S A. 2012;109(31):12734–9. doi: 10.1073/pnas.1204158109 22802646

223. Dao A, Yaro AS, Diallo M, Timbine S, Huestis DL, Kassogue Y, et al. Signatures of aestivation and migration in Sahelian malaria mosquito populations. Nature. 2014;516(7531):387–90. doi: 10.1038/nature13987 25470038

Štítky
Interné lekárstvo

Článok vyšiel v časopise

PLOS Medicine


2017 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#