1,037
Views
78
CrossRef citations to date
0
Altmetric
Review

Current and Future Perspectives on Isothermal Nucleic Acid Amplification Technologies for Diagnosing Infections

ORCID Icon &
Pages 455-483 | Published online: 12 Feb 2020

References

  • Tenover FC. The role for rapid molecular diagnostic tests for infectious diseases in precision medicine. Expert Rev Precis Med Drug Dev. 2018;3(1):69–77.
  • Al-Mulla NA, Taj-Aldeen SJ, El Shafie S, Janahi M, Al-Nasser AA, Chandra P. Bacterial bloodstream infections and antimicrobial susceptibility pattern in pediatric hematology/oncology patients after anticancer chemotherapy. Infect Drug Resist. 2014;7:289–299. doi:10.2147/IDR.S7048625395866
  • Saker L, Lee K, Cannito B, Gilmore A, Campbell-Lendrum D Globalization and infectious diseases; a review of the linkages. Special Topics No 3 UNICEF/UNDP/World Bank/WHO. 2004:1–67.
  • Cheng Y-H, Lin Y-J, Chen S-C, et al. Assessing health burden risk and control effect on dengue fever infection in the southern region of Taiwan. Infect Drug Resist. 2018;11:1423–1435. doi:10.2147/IDR.S16982030233221
  • Caliendo AM, Gilbert DN, Ginocchio CC, et al. Better tests, better care: improved diagnostics for infectious diseases. Clin Infect Dis. 2013;57 Suppl 3:S139–170. doi:10.1093/cid/cit57824200831
  • Lu TC, Tsai CL, Lee CC, et al. Preventable deaths in patients admitted from emergency department. Emerg Med J. 2006;23(6):452–455. doi:10.1136/emj.2004.02231916714507
  • Abe T, Tokuda Y, Shiraishi A, et al. In-hospital mortality associated with the misdiagnosis or unidentified site of infection at admission. Crit Care. 2019;23(1):202.31171006
  • Ahmad RA, Mahendradhata Y, Utarini A, de Vlas SJ. Diagnostic delay amongst tuberculosis patients in Jogjakarta Province, Indonesia is related to the quality of services in DOTS facilities. Trop Med Int Health. 2011;16(4):412–423. doi:10.1111/tmi.2011.16.issue-421199195
  • Bojovic O, Medenica M, Zivkovic D, et al. Factors associated with patient and health system delays in diagnosis and treatment of tuberculosis in Montenegro, 2015-2016. PLoS One. 2018;13(3):e0193997. doi:10.1371/journal.pone.019399729522545
  • Frenz MB, McIntyre AS. Reducing delays in the diagnosis and treatment of Clostridium difficile diarrhoea. QJM. 2003;96(8):579–582. doi:10.1093/qjmed/hcg09812897343
  • Elshafie S, Taj-Aldeen SJ. Emerging resistant serotypes of invasive Streptococcus pneumoniae. Infect Drug Resist. 2016;9:153–160. doi:10.2147/IDR.S10241027418844
  • Hersh AL, Shapiro DJ, Pavia AT, Shah SS. Antibiotic prescribing in ambulatory pediatrics in the United States. Pediatrics. 2011;128(6):1053–1061.22065263
  • Grijalva CG, Nuorti JP, Griffin MR. Antibiotic prescription rates for acute respiratory tract infections in US ambulatory settings. JAMA. 2009;302(7):758–766. doi:10.1001/jama.2009.116319690308
  • Roumie CL, Halasa NB, Grijalva CG, et al. Trends in antibiotic prescribing for adults in the United States–1995 to 2002. J Gen Intern Med. 2005;20(8):697–702. doi:10.1111/j.1525-1497.2005.0148.x16050877
  • Soo PC, Tseng CC, Ling SR, et al. Rapid and sensitive detection of Acinetobacter baumannii using loop-mediated isothermal amplification. J Microbiol Methods. 2013;92(2):197–200. doi:10.1016/j.mimet.2012.11.02023220188
  • Notomi T, Okayama H, Masubuchi H, et al. Loop-mediated isothermal amplification of DNA. Nucleic Acids Res. 2000;28(12):E63–E63. doi:10.1093/nar/28.12.e6310871386
  • Notomi T, Mori Y, Tomita N, Kanda H. Loop-mediated isothermal amplification (LAMP): principle, features, and future prospects. J Microbiol. 2015;53(1):1–5. doi:10.1007/s12275-015-4656-925557475
  • Li J, Macdonald J, von Stetten F. Review: a comprehensive summary of a decade development of the recombinase polymerase amplification. Analyst. 2019;144(1):31–67.
  • Dhama K, Karthik K, Chakrabort S, et al. Loop-mediated isothermal amplification of DNA (LAMP): a new diagnostic tool lights the world of diagnosis of animal and human pathogens: a review. Pak J Biol Sci. 2014;17(2):151–166. doi:10.3923/pjbs.2014.151.16624783797
  • Black EM, Lowings JP, Smith J, Heaton PR, McElhinney LM. A rapid RT-PCR method to differentiate six established genotypes of rabies and rabies-related viruses using TaqMan™ technology. J Virol Methods. 2002;105(1):25–35. doi:10.1016/S0166-0934(02)00062-912176139
  • Parida M, Sannarangaiah S, Dash PK, Rao PV, Morita K. Loop mediated isothermal amplification (LAMP): a new generation of innovative gene amplification technique; perspectives in clinical diagnosis of infectious diseases. Rev Med Virol. 2008;18(6):407–421. doi:10.1002/rmv.v18:618716992
  • Compton J. Nucleic acid sequence-based amplification. Nature. 1991;350:91. doi:10.1038/350091a01706072
  • Wharam SD, Marsh P, Lloyd JS, et al. Specific detection of DNA and RNA targets using a novel isothermal nucleic acid amplification assay based on the formation of a three-way junction structure. Nucleic Acids Res. 2001;29(11):e54–e54. doi:10.1093/nar/29.11.e5411376166
  • Walker GT, Fraiser MS, Schram JL, Little MC, Nadeau JG, Malinowski DP. Strand displacement amplification—an isothermal, in vitro DNA amplification technique. Nucleic Acids Res. 1992;20(7):1691–1696. doi:10.1093/nar/20.7.16911579461
  • Wang Y, Wang Y, Ma A-J, et al. Rapid and sensitive isothermal detection of nucleic-acid sequence by multiple cross displacement amplification. Sci Rep. 2015;5:11902. doi:10.1038/srep1190226154567
  • Barreda-García S, Miranda-Castro R, de-los-Santos-Álvarez N, Miranda-Ordieres AJ, Lobo-Castañón MJ. Helicase-dependent isothermal amplification: a novel tool in the development of molecular-based analytical systems for rapid pathogen detection. Anal Bioanal Chem. 2018;410(3):679–693. doi:10.1007/s00216-017-0620-328932883
  • Nurul Najian AB, Engku Nur Syafirah EA, Ismail N, Mohamed M, Yean CY. Development of multiplex loop mediated isothermal amplification (m-LAMP) label-based gold nanoparticles lateral flow dipstick biosensor for detection of pathogenic Leptospira. Anal Chim Acta. 2016;903:142–148. doi:10.1016/j.aca.2015.11.01526709307
  • Mori Y, Kitao M, Tomita N, Notomi T. Real-time turbidimetry of LAMP reaction for quantifying template DNA. J Biochem Biophys Methods. 2004;59(2):145–157. doi:10.1016/j.jbbm.2003.12.00515163526
  • Tomita N, Mori Y, Kanda H, Notomi T. Loop-mediated isothermal amplification (LAMP) of gene sequences and simple visual detection of products. Nat Protoc. 2008;3(5):877–882. doi:10.1038/nprot.2008.5718451795
  • Mori Y, Nagamine K, Tomita N, Notomi T. Detection of loop-mediated isothermal amplification reaction by turbidity derived from magnesium pyrophosphate formation. Biochem Biophys Res Commun. 2001;289(1):150–154. doi:10.1006/bbrc.2001.592111708792
  • Abdullah J, Saffie N, Sjasri FAR, et al. Rapid detection of Salmonella Typhi by loop-mediated isothermal amplification (LAMP) method. Braz J Microbiol. 2014;45(4):1385–1391. doi:10.1590/S1517-8382201400040003225763045
  • Solanki R, Vanjari L, Ede N, Gungi A, Soory A, Vemu L. Evaluation of LAMP assay using phenotypic tests and conventional PCR for detection of blaNDM-1 and blaKPC genes among carbapenem-resistant clinical Gram-negative isolates. J Med Microbiol. 2013;62(Pt 10):1540–1544. doi:10.1099/jmm.0.059907-023800599
  • Stratakos AC, Linton M, Millington S, Grant IR. A loop-mediated isothermal amplification method for rapid direct detection and differentiation of nonpathogenic and verocytotoxigenic Escherichia coli in beef and bovine faeces. J Appl Microbiol. 2017;122(3):817–828. doi:10.1111/jam.2017.122.issue-327992094
  • Iwamoto T, Sonobe T, Hayashi K. Loop-mediated isothermal amplification for direct detection of mycobacterium tuberculosis complex, M. avium, and M. intracellulare in sputum samples. J Clin Microbiol. 2003;41(6):2616–2622. doi:10.1128/JCM.41.6.2616-2622.200312791888
  • Liu N, Zou D, Dong D, et al. Development of a multiplex loop-mediated isothermal amplification method for the simultaneous detection of Salmonella spp. and Vibrio parahaemolyticus. Sci Rep. 2017;7:45601. doi:10.1038/srep4560128349967
  • Wang Q, Zhou Y, Li S, et al. Real-time fluorescence loop mediated isothermal amplification for the detection of Acinetobacter baumannii. PLoS One. 2013;8(7):e66406. doi:10.1371/journal.pone.006640623843955
  • Hamzan NI, Fauzi FH, Taib H, Mohamad S. Simple and rapid detection of Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans by loop-mediated isothermal amplification assay. Bangladesh J Med Sci. 2018;17:3. doi:10.3329/bjms.v17i3.36995
  • Kasahara K, Ishikawa H, Sato S, Shimakawa Y, Watanabe K. Development of multiplex loop-mediated isothermal amplification assays to detect medically important yeasts in dairy products. FEMS Microbiol Lett. 2014;357(2):208–216. doi:10.1111/1574-6968.1251224965944
  • Mahony J, Chong S, Bulir D, Ruyter A, Mwawasi K, Waltho D. Multiplex loop-mediated isothermal amplification (M-LAMP) assay for the detection of influenza A/H1, A/H3 and influenza B can provide a specimen-to-result diagnosis in 40 min with single genome copy sensitivity. J Clin Virol. 2013;58(1):127–131. doi:10.1016/j.jcv.2013.06.00623827787
  • Lucchi NW, Demas A, Narayanan J, et al. Real-time fluorescence loop mediated isothermal amplification for the diagnosis of malaria. PLoS One. 2010;5(10):e13733.21060829
  • Hara-Kudo Y, Yoshino M, Kojima T, Ikedo M. Loop-mediated isothermal amplification for the rapid detection of Salmonella. FEMS Microbiol Lett. 2005;253(1):155–161. doi:10.1016/j.femsle.2005.09.03216242860
  • Ohtsuka K, Yanagawa K, Takatori K, Hara-Kudo Y. Detection of Salmonella enterica in naturally contaminated liquid eggs by loop-mediated isothermal amplification, and characterization of Salmonella isolates. Appl Environ Microbiol. 2005;71(11):6730. doi:10.1128/AEM.71.11.6730-6735.200516269703
  • Okamura M, Ohba Y, Kikuchi S, et al. Loop-mediated isothermal amplification for the rapid, sensitive, and specific detection of the O9 group of Salmonella in chickens. Vet Microbiol. 2008;132(1):197–204. doi:10.1016/j.vetmic.2008.04.02918538511
  • Hara-Kudo Y, Konishi N, Ohtsuka K, et al. Detection of Verotoxigenic Escherichia coli O157 and O26 in food by plating methods and LAMP method: a collaborative study. Int J Food Microbiol. 2008;122(1–2):156–161. doi:10.1016/j.ijfoodmicro.2007.11.07818158196
  • Yano A, Ishimaru R, Hujikata R. Rapid and sensitive detection of heat-labile I and heat-stable I enterotoxin genes of enterotoxigenic Escherichia coli by loop-mediated isothermal amplification. J Microbiol Methods. 2007;68(2):414–420. doi:10.1016/j.mimet.2006.09.02417098313
  • Hill J, Beriwal S, Chandra I, et al. Loop-mediated isothermal amplification assay for rapid detection of common strains of Escherichia coli. J Clin Microbiol. 2008;46(8):2800–2804. doi:10.1128/JCM.00152-0818550738
  • Yamazaki W, Taguchi M, Ishibashi M, et al. Development and evaluation of a loop-mediated isothermal amplification assay for rapid and simple detection of Campylobacter jejuni and Campylobacter coli. J Med Microbiol. 2008;57(4):444–451. doi:10.1099/jmm.0.47688-018349363
  • Lloyd A, Pasupuleti V, Thota P, et al. Accuracy of loop-mediated isothermal amplification for the diagnosis of Clostridium difficile infection: a systematic review. Diagn Microbiol Infect Dis. 2015;82(1):4–10. doi:10.1016/j.diagmicrobio.2015.02.00725752201
  • Massey V, Gregson DB, Chagla AH, Storey M, John MA, Hussain Z. Clinical usefulness of components of the Triage immunoassay, enzyme immunoassay for toxins A and B, and cytotoxin B tissue culture assay for the diagnosis of Clostridium difficile diarrhea. Am J Clin Pathol. 2003;119(1):45–49. doi:10.1309/U8ATL52Q60XYAVX612520696
  • Poutanen SM, Simor AE. Clostridium difficile-associated diarrhea in adults. CMAJ. 2004;171(1):51–58. doi:10.1503/cmaj.103118915238498
  • Hataoka Y, Zhang L, Mori Y, Tomita N, Notomi T, Baba Y. Analysis of specific gene by integration of isothermal amplification and electrophoresis on poly(methyl methacrylate) microchips. Anal Chem. 2004;76(13):3689–3693. doi:10.1021/ac035032u15228342
  • Mori Y, Notomi T. Loop-mediated isothermal amplification (LAMP): a rapid, accurate, and cost-effective diagnostic method for infectious diseases. J Infect Chemother. 2009;15(2):62–69. doi:10.1007/s10156-009-0669-919396514
  • Hsieh K, Mage PL, Csordas AT, Eisenstein M, Soh HT. Simultaneous elimination of carryover contamination and detection of DNA with uracil-DNA-glycosylase-supplemented loop-mediated isothermal amplification (UDG-LAMP). Chem Commun (Camb). 2014;50(28):3747–3749. doi:10.1039/c4cc00540f24577617
  • Wong YP, Othman S, Lau YL, Radu S, Chee HY. Loop-mediated isothermal amplification (LAMP): a versatile technique for detection of micro-organisms. J Appl Microbiol. 2018;124(3):626–643. doi:10.1111/jam.1364729165905
  • Watts MR, James G, Sultana Y, et al. A loop-mediated isothermal amplification (LAMP) assay for Strongyloides stercoralis in stool that uses a visual detection method with SYTO-82 fluorescent dye. Am J Trop Med Hyg. 2014;90(2):306–311. doi:10.4269/ajtmh.13-058324323513
  • Vincent M, Xu Y, Kong H. Helicase-dependent isothermal DNA amplification. EMBO Rep. 2004;5(8):795–800. doi:10.1038/sj.embor.740020015247927
  • Abdel-Monem M, Hoffmann-Berling H. Enzymic unwinding of DNA. Eur J Biochem. 1976;65(2):431–440. doi:10.1111/j.1432-1033.1976.tb10358.x133022
  • Abdel-monem M, Dürwald H, Hoffmann-Berling H. Enzymic Unwinding of DNA. Eur J Biochem. 1976;65(2):441–449. doi:10.1111/j.1432-1033.1976.tb10359.x133023
  • Zanoli LM, Spoto G. Isothermal amplification methods for the detection of nucleic acids in microfluidic devices. Biosensors. 2012;3(1):18–43. doi:10.3390/bios301001825587397
  • An L, Tang W, Ranalli TA, Kim H-J, Wytiaz J, Kong H. Characterization of a thermostable UvrD helicase and its participation in helicase-dependent amplification. J Biol Chem. 2005;280(32):28952–28958. doi:10.1074/jbc.M50309620015955821
  • Motré A, Li Y, Kong H. Enhancing helicase-dependent amplification by fusing the helicase with the DNA polymerase. Gene. 2008;420(1):17–22. doi:10.1016/j.gene.2008.04.01718556147
  • Goldmeyer J, Kong H, Tang W. Development of a novel one-tube isothermal reverse transcription thermophilic helicase-dependent amplification platform for rapid RNA detection. J Mol Diagn. 2007;9(5):639–644. doi:10.2353/jmoldx.2007.07001217975029
  • Andresen D, von Nickisch-rosenegk M, Bier FF. Helicase dependent OnChip-amplification and its use in multiplex pathogen detection. Clin Chim Acta. 2009;403(1):244–248.19302991
  • Huang S, Do J, Mahalanabis M, et al. Low cost extraction and isothermal amplification of DNA for infectious diarrhea diagnosis. PLoS One. 2013;8(3):e60059.23555883
  • Maffert P, Reverchon S, Nasser W, Rozand C, Abaibou H. New nucleic acid testing devices to diagnose infectious diseases in resource-limited settings. Eur J Clin Microbiol Infect Dis. 2017;36(10):1717–1731.28573472
  • Hall MJ, Wharam SD, Weston A, Cardy DLN, Wilson WH. Use of signal-mediated amplification of RNA technology (SMART) to detect marine cyanophage DNA. BioTechniques. 2002;32(3):604–611.11926174
  • Li J, Macdonald J. Advances in isothermal amplification: novel strategies inspired by biological processes. Biosens Bioelectron. 2015;64:196–211. doi:10.1016/j.bios.2014.08.06925218104
  • Wharam SD, Hall MJ, Wilson WH. Detection of virus mRNA within infected host cells using an isothermal nucleic acid amplification assay: marine cyanophage gene expression within Synechococcus sp. Virol J. 2007;4(1):52. doi:10.1186/1743-422X-4-5217553149
  • Levi K, Bailey C, Bennett A, Marsh P, Cardy DLN, Towner KJ. Evaluation of an isothermal signal amplification method for rapid detection of methicillin-resistant staphylococcus aureus from patient-screening swabs. J Clin Microbiol. 2003;41(7):3187.12843062
  • Yan L, Zhou J, Zheng Y, et al. Isothermal amplified detection of DNA and RNA. Mol Biosyst. 2014;10(5):970–1003. doi:10.1039/c3mb70304e24643211
  • Deiman B, Van Aarle P, Sillekens P. Characteristics and applications of Nucleic Acid Sequence-Based Amplification (NASBA). Appl Biochem Biotechnol Part B Mol Biotechnol. 2002;20(2):163–179.
  • Gill P, Ghaemi A. Nucleic acid isothermal amplification technologies—a review. Nucleosides Nucleotides Nucleic Acids. 2008;27(3):224–243. doi:10.1080/1525777070184520418260008
  • Guatelli JC, Whitfield KM, Kwoh DY, Barringer KJ, Richman DD, Gingeras TR. Isothermal, in vitro amplification of nucleic acids by a multienzyme reaction modeled after retroviral replication. Proc Natl Acad Sci U S A. 1990;87:1874–1878. doi:10.1073/pnas.87.5.18742308948
  • Jean J, Blais B, Darveau A, Fliss I. Detection of hepatitis a virus by the nucleic acid sequence-based amplification technique and comparison with reverse transcription-PCR. Appl Environ Microbiol. 2001;67(12):5593. doi:10.1128/AEM.67.12.5593-5600.200111722911
  • Samuelson A, Westmoreland D, Eccles R, Fox JD. Development and application of a new method for amplification and detection of human rhinovirus RNA. J Virol Methods. 1998;71(2):197–209. doi:10.1016/S0166-0934(98)00006-89626953
  • van Gemen B, van Beuningen R, Nabbe A, et al. A one-tube quantitative HIV-1 RNA NASBA nucleic acid amplification assay using electrochemiluminescent (ECL) labelled probes. J Virol Methods. 1994;49(2):157–167. doi:10.1016/0166-0934(94)90040-X7822457
  • Leone G, van Gemen B, Schoen CD, van Schijndel H, Kramer FR. Molecular beacon probes combined with amplification by NASBA enable homogeneous, real-time detection of RNA. Nucleic Acids Res. 1998;26(9):2150–2155. doi:10.1093/nar/26.9.21509547273
  • Loens K, Ursi D, Goossens H, Ieven M. Nucleic acid sequence-based amplification In: Walker JM, Rapley R, editors. Medical Biomethods Handbook. Totowa, NJ: Humana Press; 2005:273–291.
  • Ginocchio CC, Kemper M, Stellrecht KA, Witt DJ. Multicenter evaluation of the performance characteristics of the nuclisens HIV-1 QT assay used for quantitation of human immunodeficiency virus type 1 RNA. J Clin Microbiol. 2003;41(1):164. doi:10.1128/JCM.41.1.164-173.200312517843
  • Loens K, Ieven M, Ursi D, et al. Detection of Mycoplasma pneumoniae by real-time nucleic acid sequence-based amplification. J Clin Microbiol. 2003;41(9):4448–4450. doi:10.1128/JCM.41.9.4448-4450.200312958290
  • Damen M, Sillekens P, Cuypers HTM, Frantzen I, Melsert R. Characterization of the quantitative HCV NASBA assay. J Virol Methods. 1999;82(1):45–54. doi:10.1016/S0166-0934(99)00079-810507412
  • Loens K, Ieven M, Ursi D, et al. Improved detection of rhinoviruses by nucleic acid sequence-based amplification after nucleotide sequence determination of the 5ʹ noncoding regions of additional rhinovirus strains. J Clin Microbiol. 2003;41(5):1971–1976. doi:10.1128/JCM.41.5.1971-1976.200312734236
  • Zappacosta R, Sablone F, Pansa L, Buca D, Buca D, Analytic RS. Diagnostic performances of human papillomavirus E6/E7 mRNA test on up-to 11-year-old liquid-based cervical samples. A biobank-based longitudinal study. Int J Mol Sci. 2017;18(7):1480.
  • Zhang F, Tetali S, Wang XP, Kaplan MH, Cromme FV, Ginocchio CC. Detection of human cytomegalovirus pp67 late gene transcripts in cerebrospinal fluid of human immunodeficiency virus type 1-infected patients by nucleic acid sequence-based amplification. J Clin Microbiol. 2000;38(5):1920.10790122
  • Morré SA, Sillekens PT, Jacobs MV, et al. Monitoring of Chlamydia trachomatis infections after antibiotic treatment using RNA detection by nucleic acid sequence based amplification. MP. 1998;51(3):149–154. doi:10.1136/mp.51.3.1499850338
  • van der Meide WF, Schoone GJ, Faber WR, et al. Quantitative nucleic acid sequence-based assay as a new molecular tool for detection and quantification of Leishmania parasites in skin biopsy samples. J Clin Microbiol. 2005;43(11):5560–5566. doi:10.1128/JCM.43.11.5560-5566.200516272487
  • van der Vliet GM, Cho SN, Kampirapap K, et al. Use of NASBA RNA amplification for detection of Mycobacterium leprae in skin biopsies from untreated and treated leprosy patients. Int J Leprosy Mycobacterial Dis. 1996;64(4):396–403.
  • van Deursen PB, Gunther AW, van Riel CC, et al. A novel quantitative multiplex NASBA method: application to measuring tissue factor and CD14 mRNA levels in human monocytes. Nucleic Acids Res. 1999;27:17. doi:10.1093/nar/27.17.e15-i
  • Jean J, Blais B, Darveau A, Fliss I. Simultaneous detection and identification of hepatitis A virus and rotavirus by multiplex nucleic acid sequence-based amplification (NASBA) and microtiter plate hybridization system. J Virol Methods. 2002;105(1):123–132. doi:10.1016/s0166-0934(02)00096-412176149
  • Klerks MM, Leone GOM, Verbeek M, van den Heuvel JFJM, Schoen CD. Development of a multiplex AmpliDet RNA for the simultaneous detection of potato leafroll virus and potato virus Y in potato tubers. J Virol Methods. 2001;93(1):115–125. doi:10.1016/S0166-0934(01)00258-011311350
  • Craw P, Balachandran W. Isothermal nucleic acid amplification technologies for point-of-care diagnostics: a critical review. Lab Chip. 2012;12(14):2469–2486. doi:10.1039/c2lc40100b22592150
  • Loens K, Ursi D, Ieven M, et al. Detection of mycoplasma pneumoniae in spiked clinical samples by nucleic acid sequence-based amplification. J Clin Microbiol. 2002;40(4):1339–1345. doi:10.1128/JCM.40.4.1339-1345.200211923354
  • Piepenburg O, Williams CH, Stemple DL, Armes NA. DNA detection using recombination proteins. PLoS Biol. 2006;4(7):e204–e204. doi:10.1371/journal.pbio.004020416756388
  • Moore MD, Jaykus L-A. Recombinase polymerase amplification: a promising point-of-care detection method for enteric viruses. Future Virol. 2017;12(8):421–429. doi:10.2217/fvl-2017-0034
  • Higgins M, Ravenhall M, Ward D, et al. PrimedRPA: primer design for recombinase polymerase amplification assays. Bioinformatics. 2018;35(4):682–684.
  • Shen F, Davydova EK, Du W, Kreutz JE, Piepenburg O, Ismagilov RF. Digital isothermal quantification of nucleic acids via simultaneous chemical initiation of recombinase polymerase amplification reactions on SlipChip. Anal Chem. 2011;83(9):3533–3540. doi:10.1021/ac200247e21476587
  • Tsaloglou MN, Watson RJ, Rushworth CM, et al. Real-time microfluidic recombinase polymerase amplification for the toxin B gene of clostridium difficile on a SlipChip platform. Analyst. 2015;140(1):258–264. doi:10.1039/C4AN01683A25371968
  • Yeh EC, Fu CC, Hu L, Thakur R, Feng J, Lee LP. Self-powered integrated microfluidic point-of-care low-cost enabling (SIMPLE) chip. Sci Adv. 2017;3(3):e1501645. doi:10.1126/sciadv.150164528345028
  • Santiago-Felipe S, Tortajada-Genaro LA, Morais S, Puchades R, Maquieira A. Isothermal DNA amplification strategies for duplex microorganism detection. Food Chem. 2015;174:509–515. doi:10.1016/j.foodchem.2014.11.08025529713
  • Liu HB, Du XJ, Zang YX, Li P, Wang S. SERS-based lateral flow strip biosensor for simultaneous detection of listeria monocytogenes and salmonella enterica serotype enteritidis. J Agric Food Chem. 2017;65(47):10290–10299. doi:10.1021/acs.jafc.7b0395729095602
  • Daher RK, Stewart G, Boissinot M, Bergeron MG. Isothermal recombinase polymerase amplification assay applied to the detection of group B streptococci in vaginal/anal samples. Clin Chem. 2014;60(4):660–666. doi:10.1373/clinchem.2013.21350424463560
  • Crannell Z, Castellanos-Gonzalez A, Nair G, Mejia R, White AC, Richards-Kortum R. Multiplexed recombinase polymerase amplification assay to detect intestinal protozoa. Anal Chem. 2016;88(3):1610–1616. doi:10.1021/acs.analchem.5b0326726669715
  • Lau HY, Wang Y, Wee EJH, Botella JR, Trau M. Field demonstration of a multiplexed point-of-care diagnostic platform for plant pathogens. Anal Chem. 2016;88(16):8074–8081. doi:10.1021/acs.analchem.6b0155127403651
  • Abd El Wahed A, Patel P, Faye O, et al. Recombinase Polymerase Amplification Assay for Rapid Diagnostics of Dengue Infection. PLoS One. 2015;10(6):e0129682. doi:10.1371/journal.pone.012968226075598
  • Abd El Wahed A, Weidmann M, Hufert FT. Diagnostics-in-a-suitcase: development of a portable and rapid assay for the detection of the emerging avian influenza A (H7N9) virus. J Clin Virol. 2015;69:16–21. doi:10.1016/j.jcv.2015.05.00426209370
  • Renner LD, Zan J, Hu LI, et al. Detection of ESKAPE bacterial pathogens at the point of care using isothermal DNA-based assays in a portable degas-actuated microfluidic diagnostic assay platform. Appl Environ Microbiol. 2017;83:4. doi:10.1128/AEM.02449-16
  • Branavan M, Mackay RE, Craw P, et al. Modular development of a prototype point of care molecular diagnostic platform for sexually transmitted infections. Med Eng Phys. 2016;38(8):741–748. doi:10.1016/j.medengphy.2016.04.02227238759
  • Hu C, Kalsi S, Zeimpekis I, et al. Ultra-fast electronic detection of antimicrobial resistance genes using isothermal amplification and Thin Film Transistor sensors. Biosens Bioelectron. 2017;96:281–287. doi:10.1016/j.bios.2017.05.01628505562
  • Kim TH, Park J, Kim CJ, Cho YK. Fully integrated lab-on-a-disc for nucleic acid analysis of food-borne pathogens. Anal Chem. 2014;86(8):3841–3848. doi:10.1021/ac403971h24635032
  • Choi G, Jung JH, Park BH, et al. A centrifugal direct recombinase polymerase amplification (direct-RPA) microdevice for multiplex and real-time identification of food poisoning bacteria. Lab Chip. 2016;16(12):2309–2316. doi:10.1039/C6LC00329J27216297
  • Lutz S, Weber P, Focke M, et al. Microfluidic lab-on-a-foil for nucleic acid analysis based on isothermal recombinase polymerase amplification (RPA). Lab Chip. 2010;10(7):887–893. doi:10.1039/b921140c20300675
  • Euler M, Wang Y, Otto P, et al. Recombinase polymerase amplification assay for rapid detection of Francisella tularensis. J Clin Microbiol. 2012;50(7):2234.22518861
  • Moore MD, Jaykus L-A. Development of a recombinase polymerase amplification assay for detection of epidemic human noroviruses. Sci Rep. 2017;7:40244. doi:10.1038/srep4024428067278
  • Yang H-L, Wei S, Gooneratne R, et al. Development of a recombinase polymerase amplification assay for Vibrio parahaemolyticus detection with an internal amplification control. Can J Microbiol. 2018;64(4):223–230. doi:10.1139/cjm-2017-050429351385
  • Ahn H, Batule BS, Seok Y, Kim M-G. Single-step recombinase polymerase amplification assay based on a paper chip for simultaneous detection of multiple foodborne pathogens. Anal Chem. 2018;90(17):10211–10216. doi:10.1021/acs.analchem.8b0130930075080
  • Qi Y, Yin Q, Shao Y, et al. Rapid and visual detection of coxiella burnetii using recombinase polymerase amplification combined with lateral flow strips. Biomed Res Int. 2018;2018:10.
  • Wang H, Sun M, Xu D, et al. Rapid visual detection of cyprinid herpesvirus 2 by recombinase polymerase amplification combined with a lateral flow dipstick. J Fish Dis. 2018;41(8):1201–1206.
  • Rohrman BA, Richards-Kortum RR. A paper and plastic device for performing recombinase polymerase amplification of HIV DNA. Lab Chip. 2012;12(17):3082–3088. doi:10.1039/c2lc40423k22733333
  • Ali MM, Li F, Zhang Z, et al. Rolling circle amplification: a versatile tool for chemical biology, materials science and medicine. Chem Soc Rev. 2014;43(10):3324–3341.24643375
  • Ali MM, Li Y. Colorimetric sensing by using allosteric-DNAzyme-coupled rolling circle amplification and a peptide nucleic acid–organic dye probe. Angew Chem Int Ed. 2009;48(19):3512–3515.
  • Zhao W, Gao Y, Kandadai SA, Brook MA, Li Y. DNA polymerization on gold nanoparticles through rolling circle amplification: towards novel scaffolds for three-dimensional periodic nanoassemblies. Angew Chem. 2006;45(15):2409–2413. doi:10.1002/anie.20060006116526071
  • Hamidi SV, Ghourchian H, Tavoosidana G. Real-time detection of H5N1 influenza virus through hyperbranched rolling circle amplification. Analyst. 2015;140(5):1502–1509. doi:10.1039/c4an01954g25627866
  • Ali MM, Su S, Filipe CDM, Pelton R, Li Y, Enzymatic manipulations of DNA oligonucleotides on microgel: towards development of DNA–microgel bioassays. Chem Commun. 2007;43:4459–4461. doi:10.1039/b709817k
  • Linck L, Reiß E, Bier F, Resch-Genger U. Direct labeling rolling circle amplification as a straightforward signal amplification technique for biodetection formats. Anal Methods. 2012;4(5):1215–1220.
  • Göransson J, Ke R, Nong RY, et al. Rapid identification of bio-molecules applied for detection of biosecurity agents using rolling circle amplification. PLoS One. 2012;7(2):e31068.22383994
  • Sato K, Ishii R, Sasaki N, Sato K, Nilsson M. Bead-based padlock rolling circle amplification for single DNA molecule counting. Anal Biochem. 2013;437(1):43–45.23467098
  • Ebai T, Souza de Oliveira FM, Löf L, et al. Analytically sensitive protein detection in microtiter plates by proximity ligation with rolling circle amplification. Clin Chem. 2017;63(9):1497. doi:10.1373/clinchem.2017.27183328667186
  • Zhang B, Wang Q, Wu J, Chen Y, Wang J. Detection of nucleic acids with a novel stem-loop primer rolling circle amplification technique. Reports. 2018;64(2):69–80.
  • Schweitzer B, Wiltshire S, Lambert J, et al. Immunoassays with rolling circle DNA amplification: a versatile platform for ultrasensitive antigen detection. Proc Natl Acad Sci. 2000;97(18):10113. doi:10.1073/pnas.17023719710954739
  • Cheng W, Yan F, Ding L, Ju H, Yin Y. Cascade signal amplification strategy for subattomolar protein detection by rolling circle amplification and quantum dots tagging. Anal Chem. 2010;82(8):3337–3342. doi:10.1021/ac100144g20345087
  • Nilsson M, Malmgren H, Samiotaki M, Kwiatkowski M, Chowdhary BP, Landegren U. Padlock probes: circularizing oligonucleotides for localized DNA detection. Science (New York, NY). 1994;265(5181):2085–2088. doi:10.1126/science.7522346
  • Polidoros AN, Pasentsis K, Tsaftaris AS. Rolling circle amplification-RACE: a method for simultaneous isolation of 5ʹ and 3ʹ cDNA ends from amplified cDNA templates. Biotechniques. 2006;41(1):35–36, 38, 40 passim. doi:10.2144/00011220516869511
  • Rubin E, Rumney S, Wang S, Kool ET. Convergent DNA synthesis: a non-enzymatic dimerization approach to circular oligodeoxynucleotides. Nucleic Acids Res. 1995;23(17):3547–3553. doi:10.1093/nar/23.17.35477567468
  • Dean FB, Nelson JR, Giesler TL, Lasken RS. Rapid amplification of plasmid and phage DNA using Phi29 DNA polymerase and multiply-primed rolling circle amplification. Genome Res. 2001;11(6):1095–1099. doi:10.1101/gr.18050111381035
  • Lizardi PM, Huang X, Zhu Z, Bray-Ward P, Thomas DC, Ward DC. Mutation detection and single-molecule counting using isothermal rolling-circle amplification. Nat Genet. 1998;19(3):225–232. doi:10.1038/8989662393
  • Zhang DY, Brandwein M, Hsuih T, Li HB. Ramification amplification: a novel isothermal DNA amplification method. Mole Diagn. 2001;6(2):141–150. doi:10.2165/00066982-200106020-00010
  • Thomas DC, Nardone GA, Randall SK. Amplification of padlock probes for DNA diagnostics by cascade rolling circle amplification or the polymerase chain reaction. Arch Pathol Lab Med. 1999;123(12):1170–1176. doi:10.1043/1543-2165-123.20.117010583921
  • Konry T, Smolina I, Yarmush JM, Irimia D, Yarmush ML. Ultrasensitive detection of low-abundance surface-marker protein using isothermal rolling circle amplification in a microfluidic nanoliter platform. Small. 2011;7(3):395–400. doi:10.1002/smll.20100162021294269
  • Zhou H, Bouwman K, Schotanus M, et al. Two-color, rolling-circle amplification on antibody microarrays for sensitive, multiplexed serum-protein measurements. Genome Biol. 2004;5(4):R28. doi:10.1186/gb-2004-5-4-r2815059261
  • Juul S, Nielsen CJ, Labouriau R, et al. Droplet microfluidics platform for highly sensitive and quantitative detection of malaria-causing plasmodium parasites based on enzyme activity measurement. ACS Nano. 2012;6(12):10676–10683. doi:10.1021/nn303859423121492
  • Ibarra-Meneses AV, Cruz I, Chicharro C, et al. Evaluation of fluorimetry and direct visualization to interpret results of a loop-mediated isothermal amplification kit to detect Leishmania DNA. Parasit Vectors. 2018;11(1):250. doi:10.1186/s13071-018-2836-229665825
  • Kuhn H, Demidov VV, Frank-Kamenetskii MD. Rolling-circle amplification under topological constraints. Nucleic Acids Res. 2002;30(2):574–580. doi:10.1093/nar/30.2.57411788721
  • Walker GT, Little MC, Nadeau JG, Shank DD. Isothermal in vitro amplification of DNA by a restriction enzyme/DNA polymerase system. Proc Natl Acad Sci. 1992;89(1):392. doi:10.1073/pnas.89.1.3921309614
  • Hellyer TJ, DesJardin LE, Teixeira L, Perkins MD, Cave MD, Eisenach KD. Detection of viable mycobacterium tuberculosis by reverse transcriptase-strand displacement amplification of mRNA. J Clin Microbiol. 1999;37(3):518–523.9986805
  • Little MC, Andrews J, Moore R, et al. Strand displacement amplification and homogeneous real-time detection incorporated in a second-generation DNA probe system, BDProbeTecET. Clin Chem. 1999;45(6):I):777–784.
  • Chen Q, Bian Z, Chen M, et al. Real-time monitoring of the strand displacement amplification (SDA) of human cytomegalovirus by a new SDA-piezoelectric DNA sensor system. Biosens Bioelectron. 2009;24(12):3412–3418. doi:10.1016/j.bios.2009.06.01219576755
  • Dong H, Zhang J, Ju H, et al. Highly sensitive multiple microRNA detection based on fluorescence quenching of graphene oxide and isothermal strand-displacement polymerase reaction. Anal Chem. 2012;84(10):4587–4593. doi:10.1021/ac300721u22510208
  • Zhang -Z-Z, Zhang C-Y. Highly sensitive detection of protein with aptamer-based target-triggering two-stage amplification. Anal Chem. 2012;84(3):1623–1629. doi:10.1021/ac202900222224936
  • Cui W, Wang L, Jiang W. A dual amplification fluorescent strategy for sensitive detection of DNA methyltransferase activity based on strand displacement amplification and DNAzyme amplification. Biosens Bioelectron. 2016;77:650–655. doi:10.1016/j.bios.2015.10.04026492469
  • Yan X, Tang M, Yang J, et al. A one-step fluorescent biosensing strategy for highly sensitive detection of HIV-related DNA based on strand displacement amplification and DNAzymes. RSC Adv. 2018;8(55):31710–31716. doi:10.1039/C8RA06480F
  • Verkooyen RP, Noordhoek GT, Klapper PE, et al. Reliability of nucleic acid amplification methods for detection of Chlamydia trachomatis in urine: results of the first international collaborative quality control study among 96 laboratories. J Clin Microbiol. 2003;41(7):3013. doi:10.1128/JCM.41.7.3013-3016.200312843035
  • Akduman D, Ehret JM, Messina K, Ragsdale S, Judson FN. Evaluation of a strand displacement amplification assay (BD ProbeTec-SDA) for detection of Neisseria gonorrhoeae in urine specimens. J Clin Microbiol. 2002;40(1):281. doi:10.1128/JCM.40.1.281-283.200211773133
  • Van Der Pol B, Ferrero DV, Buck-Barrington L, et al. Multicenter evaluation of the BDProbeTec ET System for detection of Chlamydia trachomatis and Neisseria gonorrhoeae in urine specimens, female endocervical swabs, and male urethral swabs. J Clin Microbiol. 2001;39(3):1008–1016. doi:10.1128/JCM.39.3.1008-1016.200111230419
  • Shi C, Liu Q, Ma C, Zhong W. Exponential strand-displacement amplification for detection of MicroRNAs. Anal Chem. 2014;86(1):336–339. doi:10.1021/ac403804324345199
  • Wang Y, Wang Y, Wang H, Xu J, Ye C. A label-free technique for accurate detection of nucleic acid-based self-avoiding molecular recognition systems supplemented multiple cross-displacement amplification and nanoparticles based biosensor. Artif Cells Nanomed Biotechnol. 2018;46(8):1671–1684. doi:10.1080/21691401.2017.138974829037087
  • Wang Y, Yan W, Fu S, et al. Multiple cross displacement amplification coupled with nanoparticles-based lateral flow biosensor for detection of staphylococcus aureus and identification of methicillin-resistant S. aureus. Front Microbiol. 2018;9:907. doi:10.3389/fmicb.2018.0090729867818
  • Wang Y, Li H, Li D, et al. Multiple cross displacement amplification combined with gold nanoparticle-based lateral flow biosensor for detection of vibrio parahaemolyticus. Front Microbiol. 2016;7:2047.28066368
  • Wang Y, Wang Y, Xu J, Ye C. Development of multiple cross displacement amplification label-based gold nanoparticles lateral flow biosensor for detection of Shigella spp. Front Microbiol. 2016;2016(7):1834.
  • Wang Y, Yan W, Wang Y, Xu J, Ye C. Rapid, sensitive and reliable detection of Klebsiella pneumoniae by label-free multiple cross displacement amplification coupled with nanoparticles-based biosensor. J Microbiol Methods. 2018;149:80–88. doi:10.1016/j.mimet.2018.05.00329730325
  • Wang Y, Li H, Wang Y, et al. Development of multiple cross displacement amplification label-based gold nanoparticles lateral flow biosensor for detection of Listeria monocytogenes. Int J Nanomedicine. 2017;12:473–486. doi:10.2147/IJN.S12362528138243
  • Niu L, Zhao F, Chen J, et al. Isothermal amplification and rapid detection of Klebsiella pneumoniae based on the multiple cross displacement amplification (MCDA) and gold nanoparticle lateral flow biosensor (LFB). PLoS One. 2018;13(10):e0204332. doi:10.1371/journal.pone.020433230273362
  • Asiello PJ, Baeumner AJ. Miniaturized isothermal nucleic acid amplification, a review. Lab Chip. 2011;11(8):1420–1430. doi:10.1039/c0lc00666a21387067
  • Zanoli LM, Spoto G. Isothermal amplification methods for the detection of nucleic acids in microfluidic devices. Biosensors. 2013;3(1):18. doi:10.3390/bios301001825587397
  • Esch MB, Locascio LE, Tarlov MJ, Durst RA. Detection of viable Cryptosporidium parvum using DNA-modified liposomes in a microfluidic chip. Anal Chem. 2001;73(13):2952–2958. doi:10.1021/ac001508n11467540
  • Kaarj K, Akarapipad P, Yoon J-Y. Simpler, faster, and sensitive zika virus assay using smartphone detection of loop-mediated isothermal amplification on paper microfluidic chips. Sci Rep. 2018;8(1):12438. doi:10.1038/s41598-018-30797-930127503
  • Magro L, Jacquelin B, Escadafal C, et al. Paper-based RNA detection and multiplexed analysis for Ebola virus diagnostics. Sci Rep. 2017;7(1):1347. doi:10.1038/s41598-017-00758-928465576
  • Linnes JC, Rodriguez NM, Liu L, Klapperich CM. Polyethersulfone improves isothermal nucleic acid amplification compared to current paper-based diagnostics. Biomed Microdevices. 2016;18(2):30. doi:10.1007/s10544-016-0057-z26906904
  • Becker H, Gartner C. Polymer microfabrication technologies for microfluidic systems. Anal Bioanal Chem. 2008;390(1):89–111. doi:10.1007/s00216-007-1692-217989961
  • Van Ness J, Van Ness LK, Galas DJ. Isothermal reactions for the amplification of oligonucleotides. Proc Natl Acad Sci. 2003;100(8):4504. doi:10.1073/pnas.073081110012679520
  • Kurn N, Chen P, Heath JD, Kopf-Sill A, Stephens KM, Wang S. Novel isothermal, linear nucleic acid amplification systems for highly multiplexed applications. Clin Chem. 2005;51(10):1973. doi:10.1373/clinchem.2005.05369416123149
  • Shimada M, Hino F, Sagawa H, Mukai H, Asada K, Kato I. [Development of the detection system for Mycobacterium tuberculosis DNA by using the isothermal DNA amplification method ICAN]. Rinsho Byori. 2002;50(5):528–532.12078054
  • Ma C, Liu S, Shi C. Ultrasensitive detection of microRNAs based on hairpin fluorescence probe assisted isothermal amplification. Biosens Bioelectron. 2014;58:57–60. doi:10.1016/j.bios.2014.02.03424613970
  • Connolly AR, Trau M. Rapid DNA detection by beacon-assisted detection amplification. Nat Protoc. 2011;6:772. doi:10.1038/nprot.2011.32621637197
  • Norén T, Unemo M, Magnusson C, Eiserman M, Matussek A. Evaluation of the rapid loop-mediated isothermal amplification assay illumigene for diagnosis of clostridium difficile in an outbreak situation. APMIS. 2014;122(2):155–160. doi:10.1111/apm.1212123758095
  • Lalande V, Barrault L, Wadel S, Eckert C, Petit J-C, Barbut F. Evaluation of a loop-mediated isothermal amplification assay for diagnosis of clostridium difficile infections. J Clin Microbiol. 2011;49(7):2714. doi:10.1128/JCM.01835-1021525213
  • Chow WH, McCloskey C, Tong Y, et al. Application of isothermal helicase-dependent amplification with a disposable detection device in a simple sensitive stool test for toxigenic Clostridium difficile. J Mol Diagn. 2008;10(5):452–458. doi:10.2353/jmoldx.2008.08000818669881
  • Gaydos CA, Schwebke J, Dombrowski J, et al. Clinical performance of the Solana(R) point-of-care trichomonas assay from clinician-collected vaginal swabs and urine specimens from symptomatic and asymptomatic women. Expert Rev Mol Diagn. 2017;17(3):303–306. doi:10.1080/14737159.2017.128282328092466
  • Uphoff TS, Buchan BW, Ledeboer NA, Granato PA, Daly JA, Marti TN. Multicenter evaluation of the solana group a streptococcus assay: comparison with culture. J Clin Microbiol. 2016;54(9):2388–2390. doi:10.1128/JCM.01268-1627358464
  • Goldmeyer J, Li H, McCormac M, et al. Identification of Staphylococcus aureus and determination of methicillin resistance directly from positive blood cultures by isothermal amplification and a disposable detection device. J Clin Microbiol. 2008;46(4):1534. doi:10.1128/JCM.02234-0718234878
  • Moosavian M, Seyed-Mohammadi S, Saki M, et al. Loop-mediated isothermal amplification for detection of Legionella pneumophila in respiratory specimens of hospitalized patients in Ahvaz, southwest Iran. Infect Drug Resist. 2019;12:529–534. doi:10.2147/IDR.S19809930881058
  • Couturier B, Schlaberg R, Konzak C, Nicholes J, Law C, She RC. tcdA as a diagnostic target in a loop‐mediated amplification assay for detecting toxigenic clostridium difficile. J Clin Lab Anal. 2013;27(3):171–176. doi:10.1002/jcla.2013.27.issue-323686774
  • Cheaveau J, Nguyen H, Chow B, et al. Clinical validation of a commercial LAMP test for ruling out malaria in returning travelers: a prospective diagnostic trial. Open Forum Infect Dis. 2018;5(11):ofy260–ofy260. doi:10.1093/ofid/ofy209.17230465012
  • Abirami N, Nidaullah H, Chuah L-O, et al. Evaluation of commercial loop-mediated isothermal amplification based kit and ready-to-use plating system for detection of Salmonella in naturally contaminated poultry and their processing environment. Food Control. 2016;70:74–78. doi:10.1016/j.foodcont.2016.05.035
  • Lim HSY, Zheng Q, Miks-Krajnik M, Turner M, Yuk H-G. Evaluation of commercial kit based on loop-mediated isothermal amplification for rapid detection of low levels of uninjured and injured salmonella on duck meat, bean sprouts, and fishballs in Singapore. J Food Prot. 2015;78(6):1203–1207. doi:10.4315/0362-028X.JFP-14-53526038914
  • Tegegne B, Getie S, Lemma W, Mohon AN, Pillai DR. Performance of loop-mediated isothermal amplification (LAMP) for the diagnosis of malaria among malaria suspected pregnant women in Northwest Ethiopia. Malar J. 2017;16(1):34. doi:10.1186/s12936-017-1692-428103883
  • Nguyen VAT, Nguyen HV, Dinh TV, et al. Evaluation of Loopamp™MTBC detection kit for diagnosis of pulmonary tuberculosis at a peripheral laboratory in a high burden setting. Diagn Microbiol Infect Dis. 2018;90(3):190–195. doi:10.1016/j.diagmicrobio.2017.11.00929246776
  • Matthias K, Sven E, Ulrich V, et al. Value of the eazyplex® CSF direct assay in rapid diagnosis of invasive meningococcal disease – case report. Acta Microbiol Immunol Hung. 2018;65(3):309–315. doi:10.1556/030.65.2018.02129512393
  • Findlay J, Hopkins KL, Meunier D, Woodford N. Evaluation of three commercial assays for rapid detection of genes encoding clinically relevant carbapenemases in cultured bacteria. J Antimicrob Chemother. 2015;70(5):1338–1342. doi:10.1093/jac/dku57125630646
  • Hinic V, Ziegler J, Straub C, Goldenberger D, Frei R. Extended-spectrum beta-lactamase (ESBL) detection directly from urine samples with the rapid isothermal amplification-based eazyplex(R) SuperBug CRE assay: proof of concept. J Microbiol Methods. 2015;119:203–205. doi:10.1016/j.mimet.2015.10.01526506282
  • Rodel J, Bohnert JA, Stoll S, et al. Evaluation of loop-mediated isothermal amplification for the rapid identification of bacteria and resistance determinants in positive blood cultures. Eur J Clin Microbiol Infect Dis. 2017;36(6):1033–1040. doi:10.1007/s10096-016-2888-128063000
  • Mourez T, Delaugerre C, Vray M, Lemee V, Simon F, Plantier JC. Comparison of the bioMerieux NucliSENS EasyQ HIV-1 v2.0-HIV-1 RNA quantification assay versus abbott realtime HIV-1 and Roche Cobas TaqMan HIV-1 v2.0 on current epidemic HIV-1 variants. J Clin Virol. 2015;71:76–81. doi:10.1016/j.jcv.2015.08.00726318912
  • Van Der Pol B, Hook EWI, Williams JA, Smith B, Taylor SN. Performance of the BD CTQx and GCQx amplified assays on the BD viper LT compared with the BD viper XTR system. Sex Transm Dis. 2015;42(9):521–523. doi:10.1097/OLQ.000000000000031326270114
  • Van Der Pol B, Williams JA, Fuller D, Taylor SN, Hook EW 3rd. Combined testing for chlamydia, gonorrhea, and trichomonas by use of the BD Max CT/GC/TV assay with genitourinary specimen types. J Clin Microbiol. 2017;55(1):155–164. doi:10.1128/JCM.01766-1627795343
  • Nye MB, Osiecki J, Lewinski M, et al. Detection of Chlamydia trachomatis and Neisseria gonorrhoeae with the cobas CT/NG v2.0 test: performance compared with the BD probeTec CT Qx and GC Qx amplified DNA and aptima AC2 assays. Sex Transm Infect. 2019;95(2):87. doi:10.1136/sextrans-2018-05354530126947
  • Chevaliez S, Dubernet F, Dauvillier C, Hezode C, Pawlotsky JM. The new aptima HCV quant dx real-time TMA assay accurately quantifies hepatitis C virus genotype 1-6 RNA. J Clin Virol. 2017;91:5–11. doi:10.1016/j.jcv.2017.03.02028380393
  • Bruni MP, Freitas da Silveira M, Stauffert D, et al. Aptima Trichomonas vaginalis assay elucidates significant underdiagnosis of trichomoniasis among women in Brazil according to an observational study. Sex Transm Infect. 2019;95(2):129. doi:10.1136/sextrans-2018-05356730154157
  • Cook DA, Smith LW, Law J, et al. Aptima HPV assay versus hybrid capture® 2 HPV test for primary cervical cancer screening in the HPV FOCAL trial. J Clin Virol. 2017;87:23–29. doi:10.1016/j.jcv.2016.12.00427988420
  • Ferrieri P, Nelson K, Thonen-Kerr E, Arbefeville S. Prospective evaluation of xpert xpress strep a automated PCR assay vs solana group a streptococcal NAAT vs conventional throat culture. Am J Clin Pathol. 2018;150(suppl_1):S157–S157.
  • Kurosaki Y, Magassouba NF, Oloniniyi OK, et al. Development and evaluation of Reverse Transcription-Loop-Mediated Isothermal Amplification (RT-LAMP) assay coupled with a portable device for rapid diagnosis of ebola virus disease in guinea. PLoS Negl Trop Dis. 2016;10(2):e0004472–e0004472. doi:10.1371/journal.pntd.000447226900929
  • Quoc NB, Phuong NDN, Chau NNB, Linh DTP. Closed tube loop-mediated isothermal amplification assay for rapid detection of hepatitis B virus in human blood. Heliyon. 2018;4(3):e00561. doi:10.1016/j.heliyon.2018.e0056129560471
  • Craw P, Mackay RE, Naveenathayalan A, et al. A simple, low-cost platform for real-time isothermal nucleic acid amplification. Sensors (Basel). 2015;15(9):23418–23430. doi:10.3390/s15092341826389913
  • Wang Y, Zhang B, Sun Y, Liu Y, Gu Y. Loop-mediated isothermal amplification on crude DNA as a point-of-care test for the diagnosis of mycoplasma-related vaginitis during early pregnancy. Lab Med. 2017;49(1):59–67. doi:10.1093/labmed/lmx06329272448
  • Liu D, Daubendiek SL, Zillman MA, Ryan K, Kool ET. Rolling Circle DNA Synthesis: Small Circular Oligonucleotides as Efficient Templates for DNA Polymerases. J Am Chem Soc.1996;118(7):1587–1594. doi:10.1021/ja952786k20830216
  • Walker G T.. Empirical Aspects of Strand Displacement Amplification. Genome Research. 1993; 3:1–6. doi:10.1101/gr.3.1.1