1887

Abstract

Urinary tract infections are the most common bacterial infections worldwide. Infections can range from mild, recurrent (rUTI) to complicated (cUTIs), and are predominantly caused by uropathogenic (UPEC). Antibiotic therapy is important to tackle infection; however, with the continued emergence of antibiotic resistance there is an urgent need to monitor the use of effective antibiotics through better stewardship measures. Currently, clinical diagnosis of UTIs relies on empiric methods supported by laboratory testing including cellular analysis (of both human and bacterial cells), dipstick analysis and phenotypic culture. Therefore, development of novel, sensitive and specific diagnostics is an important means to rationalise antibiotic therapy in patients. This review discusses the current diagnostic landscape and highlights promising novel diagnostic technologies in development that could aid in treatment and management of antibiotic-resistant UTIs.

  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jmm/10.1099/jmm.0.001780
2023-11-15
2024-04-28
Loading full text...

Full text loading...

/deliver/fulltext/jmm/72/11/jmm001780.html?itemId=/content/journal/jmm/10.1099/jmm.0.001780&mimeType=html&fmt=ahah

References

  1. Shetty N, Tang JW, Andrews J. Infectious Disease: Pathogenesis, Prevention and Case Studies John Wiley & Sons; 2009
    [Google Scholar]
  2. UKHSA Diagnosis of urinary tract infections: quick reference guide for primary care. In Agency UHaS 2020
    [Google Scholar]
  3. Flores-Mireles AL, Walker JN, Caparon M, Hultgren SJ. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat Rev Microbiol 2015; 13:269–284 [View Article] [PubMed]
    [Google Scholar]
  4. Bailey TP. Scott’s Diagnostic Microbiology-E-Book Elsevier Health Sciences; 2015
    [Google Scholar]
  5. Rosenberger KD, Seibert A, Hormig S. Asymptomatic GBS bacteriuria during antenatal visits: to treat or not to treat?. Nurse Pract 2020; 45:18–25 [View Article] [PubMed]
    [Google Scholar]
  6. Zeng Z, Zhan J, Zhang K, Chen H, Cheng S. Global, regional, and national burden of urinary tract infections from 1990 to 2019: an analysis of the global burden of disease study 2019. World J Urol 2022; 40:755–763 [View Article] [PubMed]
    [Google Scholar]
  7. Croker R, Walker AJ, Goldacre B. Why did some practices not implement new antibiotic prescribing guidelines on urinary tract infection? A cohort study and survey in NHS England primary care. J Antimicrob Chemother 2019; 74:1125–1132 [View Article] [PubMed]
    [Google Scholar]
  8. Harding C, Rantell A, Cardozo L, Jacobson SK, Anding R et al. How can we improve investigation, prevention and treatment for recurrent urinary tract infections - ICI-RS 2018. Neurourol Urodyn 2019; 38 Suppl 5:S90–S97 [View Article] [PubMed]
    [Google Scholar]
  9. Medina M, Castillo-Pino E. An introduction to the epidemiology and burden of urinary tract infections. Ther Adv Urol 2019; 11:1756287219832172 [View Article] [PubMed]
    [Google Scholar]
  10. Hoepelman IM. Urinary tract infection in patients with diabetes mellitus. Int J Antimicrob Agents 1994; 4:113–116 [View Article] [PubMed]
    [Google Scholar]
  11. Barnett BJ, Stephens DS. Urinary tract infection: an overview. Am J Med Sci 1997; 314:245–249 [View Article] [PubMed]
    [Google Scholar]
  12. Robino L, Scavone P, Araujo L, Algorta G, Zunino P et al. Intracellular bacteria in the pathogenesis of Escherichia coli urinary tract infection in children. Clin Infect Dis 2014; 59:e158–64 [View Article] [PubMed]
    [Google Scholar]
  13. Calzi A, Grignolo S, Caviglia I, Calevo MG, Losurdo G et al. Resistance to oral antibiotics in 4569 Gram-negative rods isolated from urinary tract infection in children. Eur J Pediatr 2016; 175:1219–1225 [View Article] [PubMed]
    [Google Scholar]
  14. Stefaniuk E, Suchocka U, Bosacka K, Hryniewicz W. Etiology and antibiotic susceptibility of bacterial pathogens responsible for community-acquired urinary tract infections in Poland. Eur J Clin Microbiol Infect Dis 2016; 35:1363–1369 [View Article]
    [Google Scholar]
  15. Kauffman CA, Fisher JF, Sobel JD, Newman CA. Candida urinary tract infections--diagnosis. Clin Infect Dis 2011; 52 Suppl 6:S452–6 [View Article] [PubMed]
    [Google Scholar]
  16. Chang P-C, Hsu Y-C, Hsieh M-L, Huang S-T, Huang H-C et al. A pilot study on Trichomonas vaginalis in women with recurrent urinary tract infections. Biomed J 2016; 39:289–294 [View Article] [PubMed]
    [Google Scholar]
  17. Behzadi P, Behzadi E, Ranjbar R. Urinary tract infections and Candida albicans. Cent European J Urol 2015; 68:96–101 [View Article] [PubMed]
    [Google Scholar]
  18. Ness D, Olsburgh J. UTI in kidney transplant. World J Urol 2020; 38:81–88 [View Article] [PubMed]
    [Google Scholar]
  19. Sullivan MJ, Ulett GC. Evaluation of hematogenous spread and ascending infection in the pathogenesis of acute pyelonephritis due to group B streptococcus in mice. Microb Pathog 2020; 138:103796 [View Article] [PubMed]
    [Google Scholar]
  20. Stewart S, Robertson C, Pan J, Kennedy S, Dancer S et al. Epidemiology of healthcare-associated infection reported from a hospital-wide incidence study: considerations for infection prevention and control planning. J Hosp Infect 2021; 114:10–22 [View Article] [PubMed]
    [Google Scholar]
  21. Tandoğdu Z, Bartoletti R, Cai T, Çek M, Grabe M et al. Antimicrobial resistance in urosepsis: outcomes from the multinational, multicenter global prevalence of infections in urology (GPIU) study 2003-2013. World J Urol 2016; 34:1193–1200 [View Article] [PubMed]
    [Google Scholar]
  22. Chaudhuri RR, Henderson IR. The evolution of the Escherichia coli phylogeny. Infect Genet Evol 2012; 12:214–226 [View Article] [PubMed]
    [Google Scholar]
  23. Picard B, Garcia JS, Gouriou S, Duriez P, Brahimi N et al. The link between phylogeny and virulence in Escherichia coli extraintestinal infection. Infect Immun 1999; 67:546–553 [View Article] [PubMed]
    [Google Scholar]
  24. Bingen E, Picard B, Brahimi N, Mathy S, Desjardins P et al. Phylogenetic analysis of Escherichia coli strains causing neonatal meningitis suggests horizontal gene transfer from a predominant pool of highly virulent B2 group strains. J Infect Dis 1998; 177:642–650 [View Article] [PubMed]
    [Google Scholar]
  25. Doumith M, Day M, Ciesielczuk H, Hope R, Underwood A et al. Rapid identification of major Escherichia coli sequence types causing urinary tract and bloodstream infections. J Clin Microbiol 2015; 53:160–166 [View Article] [PubMed]
    [Google Scholar]
  26. Findlay J, Gould VC, North P, Bowker KE, Williams MO et al. Characterization of cefotaxime-resistant urinary Escherichia coli from primary care in South-West England 2017-18. J Antimicrob Chemother 2020; 75:65–71 [View Article] [PubMed]
    [Google Scholar]
  27. Johnson JR, Menard M, Johnston B, Kuskowski MA, Nichol K et al. Epidemic clonal groups of Escherichia coli as a cause of antimicrobial-resistant urinary tract infections in Canada, 2002 to 2004. Antimicrob Agents Chemother 2009; 53:2733–2739 [View Article]
    [Google Scholar]
  28. Johnson JR, Delavari P, Kuskowski M, Stell AL. Phylogenetic distribution of extraintestinal virulence-associated traits in Escherichia coli. J Infect Dis 2001; 183:78–88 [View Article] [PubMed]
    [Google Scholar]
  29. Johnson JR, Kuskowski MA, Owens K, Gajewski A, Winokur PL. Phylogenetic origin and virulence genotype in relation to resistance to fluoroquinolones and/or extended-spectrum cephalosporins and cephamycins among Escherichia coli isolates from animals and humans. J Infect Dis 2003; 188:759–768 [View Article] [PubMed]
    [Google Scholar]
  30. Johnson JR, Porter SB, Zhanel G, Kuskowski MA, Denamur E et al. Virulence of Escherichia coli clinical isolates in a murine sepsis model in relation to sequence type ST131 status, fluoroquinolone resistance, and virulence genotype. Infect Immun 2012; 80:1554–1562 [View Article]
    [Google Scholar]
  31. Lau SH, Kaufmann ME, Livermore DM, Woodford N, Willshaw GA et al. UK epidemic Escherichia coli strains A-E, with CTX-M-15 beta-lactamase, all belong to the international O25:H4-ST131 clone. J Antimicrob Chemother 2008; 62:1241–1244 [View Article] [PubMed]
    [Google Scholar]
  32. Petty NK, Ben Zakour NL, Stanton-Cook M, Skippington E, Totsika M et al. Global dissemination of a multidrug resistant Escherichia coli clone. Proc Natl Acad Sci U S A 2014; 111:5694–5699 [View Article] [PubMed]
    [Google Scholar]
  33. Pitout JDD, Gregson DB, Campbell L, Laupland KB. Molecular characteristics of extended-spectrum-beta-lactamase-producing Escherichia coli isolates causing bacteremia in the Calgary Health Region from 2000 to 2007: emergence of clone ST131 as a cause of community-acquired infections. Antimicrob Agents Chemother 2009; 53:2846–2851 [View Article] [PubMed]
    [Google Scholar]
  34. Cagnacci S, Gualco L, Debbia E, Schito GC, Marchese A. European emergence of ciprofloxacin-resistant Escherichia coli clonal groups O25:H4-ST 131 and O15:K52:H1 causing community-acquired uncomplicated cystitis. J Clin Microbiol 2008; 46:2605–2612 [View Article] [PubMed]
    [Google Scholar]
  35. Coque TM, Novais A, Carattoli A, Poirel L, Pitout J et al. Dissemination of clonally related Escherichia coli strains expressing extended-spectrum beta-lactamase CTX-M-15. Emerg Infect Dis 2008; 14:195–200 [View Article] [PubMed]
    [Google Scholar]
  36. Nicolas-Chanoine M-H, Blanco J, Leflon-Guibout V, Demarty R, Alonso MP et al. Intercontinental emergence of Escherichia coli clone O25:H4-ST131 producing CTX-M-15. J Antimicrob Chemother 2008; 61:273–281 [View Article] [PubMed]
    [Google Scholar]
  37. Morris D, McGarry E, Cotter M, Passet V, Lynch M et al. Detection of OXA-48 carbapenemase in the pandemic clone Escherichia coli O25b:H4-ST131 in the course of investigation of an outbreak of OXA-48-producing Klebsiella pneumoniae. Antimicrob Agents Chemother 2012; 56:4030–4031 [View Article] [PubMed]
    [Google Scholar]
  38. Taati Moghadam M, Mirzaei M, Fazel Tehrani Moghaddam M, Babakhani S, Yeganeh O et al. The challenge of global emergence of novel colistin-resistant Escherichia coli ST131. Microb Drug Resist 2021; 27:1513–1524 [View Article] [PubMed]
    [Google Scholar]
  39. Gupta K, Hooton TM, Naber KG, Wullt B, Colgan R et al. International clinical practice guidelines for the treatment of acute uncomplicated cystitis and pyelonephritis in women: a 2010 update by the Infectious Diseases Society of America and the European Society for Microbiology and Infectious Diseases. Clin Infect Dis 2011; 52:e103–20 [View Article] [PubMed]
    [Google Scholar]
  40. Bains A, Buna D, Hoag NA. A retrospective review assessing the efficacy and safety of nitrofurantoin in renal impairment. Can Pharm J 2009; 142:248–252 [View Article]
    [Google Scholar]
  41. UKHSA Urinary Tract Infection (Lower): Antimicrobial Prescribing Agency UHaS; 2018
    [Google Scholar]
  42. Kornfält Isberg H, Melander E, Hedin K, Mölstad S, Beckman A. Uncomplicated urinary tract infections in Swedish primary care; etiology, resistance and treatment. BMC Infect Dis 2019; 19:155 [View Article] [PubMed]
    [Google Scholar]
  43. Kranz J, Schmidt S, Lebert C, Schneidewind L, Mandraka F et al. The 2017 update of the German clinical guideline on epidemiology, diagnostics, therapy, prevention, and management of uncomplicated urinary tract infections in adult patients. Part II: therapy and prevention. Urol Int 2018; 100:271–278 [View Article]
    [Google Scholar]
  44. Piraux A, Faure S, Naber KG, Alidjanov JF, Ramond-Roquin A. Changes in the management of urinary tract infections in women: impact of the new recommendations on antibiotic prescribing behavior in France, between 2014 and 2019. BMC Health Serv Res 2021; 21:612 [View Article] [PubMed]
    [Google Scholar]
  45. CDC Adult outpatient treatment recommendations - antibiotic use; 2021 https://www.cdc.gov/antibiotic-use/clinicians/adult-treatment-rec.html accessed 25 August 2022
  46. EAU Eau guidelines on urological infections - the guideline; 2022 https://uroweb.org/guidelines/urological-infections/chapter/the-guideline accessed 25 August 2022
  47. IDSA Uncomplicated Cystitis and Pyelonephritis (UTI); 2011 https://www.idsociety.org/practice-guideline/uncomplicated-cystitis-and-pyelonephritis-uti/ accessed 20 October 2022
  48. Elias C, Moja L, Mertz D, Loeb M, Forte G et al. Guideline recommendations and antimicrobial resistance: the need for a change. BMJ Open 2017; 7:e016264 [View Article] [PubMed]
    [Google Scholar]
  49. FMHS Directorate General of Pharmacy – Sudan national standard treatment guidelines; 2014 http://www.sho.gov.sd/controller/dwn_hub_files.php?id=915 accessed 26 October 2022
  50. NCDCI National Treatment Guidelines for Antimicrobial Use in Infectious Diseases; 2016 https://ncdc.gov.in/WriteReadData/l892s/File622.pdf accessed 21 October 2022
  51. Adhikari B, Pokharel S, Raut S, Adhikari J, Thapa S et al. Why do people purchase antibiotics over-the-counter? A qualitative study with patients, clinicians and dispensers in central, eastern and western Nepal. BMJ Glob Health 2021; 6:e005829 [View Article] [PubMed]
    [Google Scholar]
  52. Do NTT, Vu HTL, Nguyen CTK, Punpuing S, Khan WA et al. Community-based antibiotic access and use in six low-income and middle-income countries: a mixed-method approach. Lancet Glob Health 2021; 9:e610–e619 [View Article] [PubMed]
    [Google Scholar]
  53. Belachew SA, Hall L, Selvey LA. Non-prescription dispensing of antibiotic agents among community drug retail outlets in Sub-Saharan African countries: a systematic review and meta-analysis. Antimicrob Resist Infect Control 2021; 10:13 [View Article] [PubMed]
    [Google Scholar]
  54. Kotwani A, Joshi J, Lamkang AS. Over-the-counter sale of antibiotics in India: a qualitative study of providers’ perspectives across two States. Antibiotics 2021; 10:1123 [View Article] [PubMed]
    [Google Scholar]
  55. Murray CJL, Ikuta KS, Sharara F, Swetschinski L, Robles Aguilar G et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. The Lancet 2022; 399:629–655 [View Article]
    [Google Scholar]
  56. Vikesland P, Garner E, Gupta S, Kang S, Maile-Moskowitz A et al. Differential drivers of antimicrobial resistance across the world. Acc Chem Res 2019; 52:916–924 [View Article] [PubMed]
    [Google Scholar]
  57. Joshi LT. The G7 Summit 2021: time for our world leaders to step up to the challenge of antimicrobial resistance. Access Microbiol 2021; 3:000298 [View Article] [PubMed]
    [Google Scholar]
  58. UKHSA Urinary Tract Infection (Recurrent): Antimicrobial Prescribing Agency UHaS; 2018
    [Google Scholar]
  59. Fisher H, Oluboyede Y, Chadwick T, Abdel-Fattah M, Brennand C et al. Continuous low-dose antibiotic prophylaxis for adults with repeated urinary tract infections (AnTIC): a randomised, open-label trial. Lancet Infect Dis 2018; 18:957–968 [View Article] [PubMed]
    [Google Scholar]
  60. Ahmed H, Farewell D, Jones HM, Francis NA, Paranjothy S et al. Antibiotic prophylaxis and clinical outcomes among older adults with recurrent urinary tract infection: cohort study. Age Ageing 2019; 48:228–234 [View Article] [PubMed]
    [Google Scholar]
  61. Jent P, Berger J, Kuhn A, Trautner BW, Atkinson A et al. Antibiotics for preventing recurrent urinary tract infection: systematic review and meta-analysis. Open Forum Infect Dis 2022; 9:ofac327 [View Article] [PubMed]
    [Google Scholar]
  62. Alsubaie SS, Barry MA. Current status of long-term antibiotic prophylaxis for urinary tract infections in children: an antibiotic stewardship challenge. Kidney Res Clin Pract 2019; 38:441–454 [View Article] [PubMed]
    [Google Scholar]
  63. Langford BJ, Brown KA, Diong C, Marchand-Austin A, Adomako K et al. The benefits and harms of antibiotic prophylaxis for urinary tract infection in older adults. Clin Infect Dis 2021; 73:e782–e791 [View Article] [PubMed]
    [Google Scholar]
  64. Care UG-DoHaS Annual Report of the Chief Medical Officer; 2011
  65. Wisell KT, Kahlmeter G, Giske CG. Trimethoprim and enterococci in urinary tract infections: new perspectives on an old issue. J Antimicrob Chemother 2008; 62:35–40 [View Article] [PubMed]
    [Google Scholar]
  66. Kahlmeter G, Åhman J, Matuschek E. Antimicrobial resistance of Escherichia coli causing uncomplicated urinary tract infections: a European update for 2014 and comparison with 2000 and 2008. Infect Dis Ther 2015; 4:417–423 [View Article] [PubMed]
    [Google Scholar]
  67. McNulty CAM, Lasseter GM, Charlett A, Lovering A, Howell-Jones R et al. Does laboratory antibiotic susceptibility reporting influence primary care prescribing in urinary tract infection and other infections?. J Antimicrob Chemother 2011; 66:1396–1404 [View Article] [PubMed]
    [Google Scholar]
  68. Somorin YM, Weir N-JM, Pattison SH, Crockard MA, Hughes CM et al. Antimicrobial resistance in urinary pathogens and culture-independent detection of trimethoprim resistance in urine from patients with urinary tract infection. BMC Microbiol 2022; 22:144 [View Article] [PubMed]
    [Google Scholar]
  69. Sloane PD, Kistler CE, Reed D, Weber DJ, Ward K et al. Urine culture testing in community nursing homes: gateway to antibiotic overprescribing. Infect Control Hosp Epidemiol 2017; 38:524–531 [View Article] [PubMed]
    [Google Scholar]
  70. Nicolle LE. Antimicrobial stewardship in long term care facilities: what is effective?. Antimicrob Resist Infect Control 2014; 3:6 [View Article] [PubMed]
    [Google Scholar]
  71. Zabarsky TF, Sethi AK, Donskey CJ. Sustained reduction in inappropriate treatment of asymptomatic bacteriuria in a long-term care facility through an educational intervention. Am J Infect Control 2008; 36:476–480 [View Article] [PubMed]
    [Google Scholar]
  72. Woodford HJ, George J. Diagnosis and management of urinary tract infection in hospitalized older people. J Am Geriatr Soc 2009; 57:107–114 [View Article] [PubMed]
    [Google Scholar]
  73. Rousham E, Cooper M, Petherick E, Saukko P, Oppenheim B. Overprescribing antibiotics for asymptomatic bacteriuria in older adults: a case series review of admissions in two UK hospitals. Antimicrob Resist Infect Control 2019; 8:71 [View Article] [PubMed]
    [Google Scholar]
  74. Phillips CD, Adepoju O, Stone N, Moudouni DKM, Nwaiwu O et al. Asymptomatic bacteriuria, antibiotic use, and suspected urinary tract infections in four nursing homes. BMC Geriatr 2012; 12:73 [View Article] [PubMed]
    [Google Scholar]
  75. D’Agata E, Loeb MB, Mitchell SL. Challenges in assessing nursing home residents with advanced dementia for suspected urinary tract infections. J Am Geriatr Soc 2013; 61:62–66 [View Article] [PubMed]
    [Google Scholar]
  76. Joseph A, McGowan T, Weston V, Ogunbuyide O, Bird S et al. 130“to dip or not to dip”: a quality improvement project to improve the diagnosis and management of urinary tract infection in care homes. Age and Ageing 2018; 47:iii31–iii42 [View Article]
    [Google Scholar]
  77. Shallcross LJ, Rockenschaub P, McNulty D, Freemantle N, Hayward A et al. Diagnostic uncertainty and urinary tract infection in the emergency department: a cohort study from a UK hospital. BMC Emerg Med 2020; 20:40 [View Article] [PubMed]
    [Google Scholar]
  78. Goering RV. Mims’ Medical Microbiology and Immunology Elsevier; 2020
    [Google Scholar]
  79. Musher DM, Thorsteinsson SB, Airola VM II. Quantitative urinalysis. Diagnosing urinary tract infection in men. JAMA 1976; 236:2069–2072 [View Article] [PubMed]
    [Google Scholar]
  80. Ditchburn RK, Ditchburn JS. A study of microscopical and chemical tests for the rapid diagnosis of urinary tract infections in general practice. Br J Gen Pract 1990; 40:406–408 [PubMed]
    [Google Scholar]
  81. Vickers D, Ahmad T, Coulthard MG. Diagnosis of urinary tract infection in children: fresh urine microscopy or culture?. Lancet 1991; 338:767–770 [View Article] [PubMed]
    [Google Scholar]
  82. Leman P. Validity of urinalysis and microscopy for detecting urinary tract infection in the emergency department. Eur J Emerg Med 2002; 9:141–147 [View Article] [PubMed]
    [Google Scholar]
  83. Wiwanitkit V, Udomsantisuk N, Boonchalermvichian C. Diagnostic value and cost utility analysis for urine Gram stain and urine microscopic examination as screening tests for urinary tract infection. Urol Res 2005; 33:220–222 [View Article] [PubMed]
    [Google Scholar]
  84. Beyer AK, Currea GCC, Holm A. Validity of microscopy for diagnosing urinary tract infection in general practice - a systematic review. Scand J Prim Health Care 2019; 37:373–379 [View Article] [PubMed]
    [Google Scholar]
  85. Dadzie I, Quansah E, Puopelle Dakorah M, Abiade V, Takyi-Amuah E et al. The effectiveness of dipstick for the detection of urinary tract infection. Can J Infect Dis Med Microbiol 2019; 2019:8642628 [View Article] [PubMed]
    [Google Scholar]
  86. Patel HD, Livsey SA, Swann RA, Bukhari SS. Can urine dipstick testing for urinary tract infection at point of care reduce laboratory workload?. J Clin Pathol 2005; 58:951–954 [View Article] [PubMed]
    [Google Scholar]
  87. Chernaya A, Søborg C, Midttun M. Validity of the urinary dipstick test in the diagnosis of urinary tract infections in adults. Dan Med J 2021; 69:A07210607 [PubMed]
    [Google Scholar]
  88. Service NH. Urinary tract infection checklist for care homes; 2018 https://www.england.nhs.uk/atlas_case_study/urinary-tract-infection-checklist-for-care-homes/ accessed 4 July 2023
  89. Perry JL, Matthews JS, Weesner DE. Evaluation of leukocyte esterase activity as a rapid screening technique for bacteriuria. J Clin Microbiol 1982; 15:852–854 [View Article] [PubMed]
    [Google Scholar]
  90. UKHSA UK Standards for Microbiology Investigations - Investigation of Urine Agency UHaS; 2018
    [Google Scholar]
  91. Dong Q, Nelson DE, Toh E, Diao L, Gao X et al. The microbial communities in male first catch urine are highly similar to those in paired urethral swab specimens. PLoS One 2011; 6:e19709 [View Article] [PubMed]
    [Google Scholar]
  92. Nelson DE, Van Der Pol B, Dong Q, Revanna KV, Fan B et al. Characteristic male urine microbiomes associate with asymptomatic sexually transmitted infection. PLoS One 2010; 5:e14116 [View Article] [PubMed]
    [Google Scholar]
  93. Siddiqui H, Nederbragt AJ, Lagesen K, Jeansson SL, Jakobsen KS. Assessing diversity of the female urine microbiota by high throughput sequencing of 16S rDNA amplicons. BMC Microbiol 2011; 11:244 [View Article] [PubMed]
    [Google Scholar]
  94. Joshi LT, Jones IA, Silver K. Encyclopedia of Infection and Immunity Elsevier; 2022
    [Google Scholar]
  95. KASS EH. Bacteriuria and the diagnosis of infections of the urinary tract; with observations on the use of methionine as a urinary antiseptic. AMA Arch Intern Med 1957; 100:709–714 [View Article] [PubMed]
    [Google Scholar]
  96. Kunin CM, White LV, Hua TH. A reassessment of the importance of “low-count” bacteriuria in young women with acute urinary symptoms. Ann Intern Med 1993; 119:454–460 [View Article] [PubMed]
    [Google Scholar]
  97. Tullus K. Low urinary bacterial counts: do they count?. Pediatr Nephrol 2016; 31:171–174 [View Article] [PubMed]
    [Google Scholar]
  98. Neugent ML, Hulyalkar NV, Nguyen VH, Zimmern PE, De Nisco NJ. Advances in understanding the human urinary microbiome and its potential role in urinary tract infection. mBio 2020; 11:e00218-20 [View Article] [PubMed]
    [Google Scholar]
  99. Beebout CJ, Robertson GL, Reinfeld BI, Blee AM, Morales GH et al. Uropathogenic Escherichia coli subverts mitochondrial metabolism to enable intracellular bacterial pathogenesis in urinary tract infection. Nat Microbiol 2022; 7:1348–1360 [View Article] [PubMed]
    [Google Scholar]
  100. Batchelor BI, Hunt AR, Bowler IC, Crook DW. Laboratory detection of leucocyte esterase and nitrite as an alternative to urine microscopy. Eur J Clin Microbiol Infect Dis 1996; 15:663–664 [View Article] [PubMed]
    [Google Scholar]
  101. Tran A, Alby K, Kerr A, Jones M, Gilligan PH. Cost savings realized by implementation of routine microbiological identification by matrix-assisted laser desorption ionization-time of flight mass spectrometry. J Clin Microbiol 2015; 53:2473–2479 [View Article] [PubMed]
    [Google Scholar]
  102. Veron L, Mailler S, Girard V, Muller BH, L’Hostis G et al. Rapid urine preparation prior to identification of uropathogens by MALDI-TOF MS. Eur J Clin Microbiol Infect Dis 2015; 34:1787–1795 [View Article] [PubMed]
    [Google Scholar]
  103. Ferreira L, Sánchez-Juanes F, González-Avila M, Cembrero-Fuciños D, Herrero-Hernández A et al. Direct identification of urinary tract pathogens from urine samples by matrix-assisted laser desorption ionization-time of flight mass spectrometry. J Clin Microbiol 2010; 48:2110–2115 [View Article] [PubMed]
    [Google Scholar]
  104. Zboromyrska Y, Rubio E, Alejo I, Vergara A, Mons A et al. Development of a new protocol for rapid bacterial identification and susceptibility testing directly from urine samples. Clin Microbiol Infect 2016; 22:561 [View Article] [PubMed]
    [Google Scholar]
  105. Sun C, Zhang X, Wang J, Cheng C, Kang H et al. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry combined with UF-5000i urine flow cytometry to directly identify pathogens in clinical urine specimens within 1 hour. Ann Transl Med 2020; 8:602 [View Article] [PubMed]
    [Google Scholar]
  106. Oviaño M, Ramírez C de la L, Barbeyto LP, Bou G. Rapid direct detection of carbapenemase-producing Enterobacteriaceae in clinical urine samples by MALDI-TOF MS analysis. J Antimicrob Chemother 2017; 72:1350–1354 [View Article] [PubMed]
    [Google Scholar]
  107. Chen C, Hong W. Recent development of rapid antimicrobial susceptibility testing methods through metabolic profiling of bacteria. Antibiotics 2021; 10:311 [View Article] [PubMed]
    [Google Scholar]
  108. Gajic I, Kabic J, Kekic D, Jovicevic M, Milenkovic M et al. Antimicrobial susceptibility testing: a comprehensive review of currently used methods. Antibiotics 2022; 11:427 [View Article] [PubMed]
    [Google Scholar]
  109. BioMérieux VITEK 2; 2022 https://www.biomerieux-diagnostics.com/vitekr-2-0 accessed 14 September 2022
  110. Becton, Dickinson, (BD) The implementation of the EUCAST standard in BD PhoenixTM and BD EpiCenterTM Systems; 2022 https://www.bd.com/resource.aspx?IDX=10841 accessed 14 September 2022
  111. Beckman C. DxM MicroScan WalkAway ID/AST; 2022 https://www.beckmancoulter.com/products/microbiology/dxm-microscan-walkaway-system accessed 15 September 2022
  112. Hernández-Durán M, López-Jácome LE, Colín-Castro CA, Cerón-González G, Ortega-Peña S et al. Comparison of the microscan walkaway and vitek 2 compact systems for the identification and susceptibility of clinical gram-positive and gram-negative bacteria. Investigación en discapacidad 2017; 6:105–114
    [Google Scholar]
  113. van Belkum A, Dunne WM Jr. Next-generation antimicrobial susceptibility testing. J Clin Microbiol 2013; 51:2018–2024 [View Article] [PubMed]
    [Google Scholar]
  114. Smieszek T, Pouwels KB, Dolk FCK, Smith DRM, Hopkins S et al. Potential for reducing inappropriate antibiotic prescribing in English primary care. J Antimicrob Chemother 2018; 73:ii36–ii43 [View Article] [PubMed]
    [Google Scholar]
  115. Cooke J, Butler C, Hopstaken R, Dryden MS, McNulty C et al. Narrative review of primary care point-of-care testing (POCT) and antibacterial use in respiratory tract infection (RTI). BMJ Open Respir Res 2015; 2:e000086 [View Article] [PubMed]
    [Google Scholar]
  116. Nora D, Salluh J, Martin-Loeches I, Póvoa P. Biomarker-guided antibiotic therapy-strengths and limitations. Ann Transl Med 2017; 5:208 [View Article] [PubMed]
    [Google Scholar]
  117. Levine AR, Tran M, Shepherd J, Naut E. Utility of initial procalcitonin values to predict urinary tract infection. Am J Emerg Med 2018; 36:1993–1997 [View Article] [PubMed]
    [Google Scholar]
  118. Ward C. Point-of-care C-reactive protein testing to optimise antibiotic use in a primary care urgent care centre setting. BMJ Open Qual 2018; 7:e000391 [View Article]
    [Google Scholar]
  119. Cooke J, Llor C, Hopstaken R, Dryden M, Butler C. Respiratory tract infections (RTIs) in primary care: narrative review of C reactive protein (CRP) point-of-care testing (POCT) and antibacterial use in patients who present with symptoms of RTI. BMJ Open Resp Res 2020; 7:e000624 [View Article]
    [Google Scholar]
  120. Kengne AP, Batty GD, Hamer M, Stamatakis E, Czernichow S. Association of C-reactive protein with cardiovascular disease mortality according to diabetes status: pooled analyses of 25,979 participants from four U.K. prospective cohort studies. Diabetes Care 2012; 35:396–403 [View Article] [PubMed]
    [Google Scholar]
  121. Pitharouli MC, Hagenaars SP, Glanville KP, Coleman JRI, Hotopf M et al. Elevated C-reactive protein in patients with depression, independent of genetic, health, and psychosocial factors: results from the UK biobank. Am J Psychiatry 2021; 178:522–529 [View Article] [PubMed]
    [Google Scholar]
  122. Fraile Navarro D, Sullivan F, Azcoaga-Lorenzo A, Hernandez Santiago V. Point-of-care tests for urinary tract infections: protocol for a systematic review and meta-analysis of diagnostic test accuracy. BMJ Open 2020; 10:e033424 [View Article]
    [Google Scholar]
  123. Xu R-Y, Liu H-W, Liu J-L, Dong J-H. Procalcitonin and C-reactive protein in urinary tract infection diagnosis. BMC Urol 2014; 14: [View Article]
    [Google Scholar]
  124. Kuil SD, Hidad S, Fischer JC, Harting J, Hertogh CMPM et al. Sensitivity of C-reactive protein and procalcitonin measured by point-of-care tests to diagnose urinary tract infections in nursing home residents: a cross-sectional study. Clin Infect Dis 2021; 73:e3867–e3875 [View Article] [PubMed]
    [Google Scholar]
  125. Moustafa A, Li W, Singh H, Moncera KJ, Torralba MG et al. Microbial metagenome of urinary tract infection. Sci Rep 2018; 8: [View Article]
    [Google Scholar]
  126. Couto N, Schuele L, Raangs EC, Machado MP, Mendes CI et al. Critical steps in clinical shotgun metagenomics for the concomitant detection and typing of microbial pathogens. Sci Rep 2018; 8:13767 [View Article] [PubMed]
    [Google Scholar]
  127. Wang C-X, Huang Z, Fang W, Zhang Z, Fang X et al. Preliminary assessment of nanopore-based metagenomic sequencing for the diagnosis of prosthetic joint infection. Int J Infect Dis 2020; 97:54–59 [View Article] [PubMed]
    [Google Scholar]
  128. Salzberg SL, Yorke JA. Beware of mis-assembled genomes. Bioinformatics 2005; 21:4320–4321 [View Article]
    [Google Scholar]
  129. Illumina Introduction to NGS; 2022 https://www.illumina.com/science/technology/next-generation-sequencing.html accessed 21 October 2022
  130. ThermoFisher, Scientific Ion Torrent; 2022 https://www.thermofisher.com/uk/en/home/brands/ion-torrent.html accessed 21 October 2022
  131. Petersen LM, Martin IW, Moschetti WE, Kershaw CM, Tsongalis GJ. Third-generation sequencing in the clinical laboratory: exploring the advantages and challenges of nanopore sequencing. J Clin Microbiol 2019; 58:e01315-19 [View Article] [PubMed]
    [Google Scholar]
  132. Bleidorn C. Third generation sequencing: technology and its potential impact on evolutionary biodiversity research. Syst Biodivers 2016; 14:1–8 [View Article]
    [Google Scholar]
  133. Oxford, Nanopore Nanopore Technology; 2022 https://nanoporetech.com accessed 22 October 2022
  134. Pacific, Bio, (PacBio) How it works; 2022 https://www.pacb.com/technology/hifi-sequencing/how-it-works/ accessed 22 October 2022
  135. Ardui S, Ameur A, Vermeesch JR, Hestand MS. Single molecule real-time (SMRT) sequencing comes of age: applications and utilities for medical diagnostics. Nucleic Acids Res 2018; 46:2159–2168 [View Article]
    [Google Scholar]
  136. Charalampous T, Kay GL, Richardson H, Aydin A, Baldan R et al. Nanopore metagenomics enables rapid clinical diagnosis of bacterial lower respiratory infection. Nat Biotechnol 2019; 37:783–792 [View Article]
    [Google Scholar]
  137. Schmidt K, Mwaigwisya S, Crossman LC, Doumith M, Munroe D et al. Identification of bacterial pathogens and antimicrobial resistance directly from clinical urines by nanopore-based metagenomic sequencing. J Antimicrob Chemother 2017; 72:104–114 [View Article] [PubMed]
    [Google Scholar]
  138. Votintseva AA, Bradley P, Pankhurst L, del Ojo Elias C, Loose M et al. Same-day diagnostic and surveillance data for tuberculosis via whole-genome sequencing of direct respiratory samples. J Clin Microbiol 2017; 55:1285–1298 [View Article]
    [Google Scholar]
  139. Břinda K, Callendrello A, Ma KC, MacFadden DR, Charalampous T et al. Rapid inference of antibiotic resistance and susceptibility by genomic neighbour typing. Nat Microbiol 2020; 5:455–464 [View Article] [PubMed]
    [Google Scholar]
  140. Zhang L, Huang W, Zhang S, Li Q, Wang Y et al. Rapid detection of bacterial pathogens and antimicrobial resistance genes in clinical urine samples with urinary tract infection by metagenomic Nanopore sequencing. Front Microbiol 2022; 13:858777 [View Article] [PubMed]
    [Google Scholar]
  141. Li M, Yang F, Lu Y, Huang W. Identification of Enterococcus faecalis in a patient with urinary-tract infection based on metagenomic next-generation sequencing: a case report. BMC Infect Dis 2020; 20: [View Article]
    [Google Scholar]
  142. Huang Y-T, Liu P-Y, Shih P-W. Homopolish: a method for the removal of systematic errors in nanopore sequencing by homologous polishing. Genome Biol 2021; 22: [View Article]
    [Google Scholar]
  143. Oxford, Nanopore Epi2me; 2022 https://epi2me.nanoporetech.com/ accessed 28 October 2022
  144. Oxford, Nanopore VolTRAX; 2022 https://nanoporetech.com/products/voltrax accessed 28 October 2022
  145. Kerkhof LJ. Is Oxford Nanopore sequencing ready for analyzing complex microbiomes?. FEMS Microbiol Ecol 2021; 97:fiab001 [View Article] [PubMed]
    [Google Scholar]
  146. Ammer-Herrmenau C, Pfisterer N, van den Berg T, Gavrilova I, Amanzada A et al. Comprehensive wet-bench and bioinformatics workflow for complex microbiota using Oxford Nanopore technologies. mSystems 2021; 6:e0075021 [View Article] [PubMed]
    [Google Scholar]
  147. Patil S, Augustine D, Sowmya S, Haragannavar VC, Gujjar N et al. Nanopore sequencing technology in oral oncology: a comprehensive insight. J Contemp Dent Pract 2022; 23:268–275 [View Article]
    [Google Scholar]
  148. Scheller FW, Wollenberger U, Warsinke A, Lisdat F. Research and development in biosensors. Curr Opin Biotechnol 2001; 12:35–40 [View Article] [PubMed]
    [Google Scholar]
  149. WHO Accessible quality-assured diagnostics; 2009 https://apps.who.int/iris/bitstream/handle/10665/84529/TDR_BL7.10_eng.pdf?sequence=1&isAllowed=y accessed 19 September 2022
  150. Turner A, Karube I, Wilson GS. eds Biosensors: Fundamentals and Applications Oxford, New York: Oxford University Press; 1987
    [Google Scholar]
  151. Gomez-Cruz J, Nair S, Manjarrez-Hernandez A, Gavilanes-Parra S, Ascanio G et al. Cost-effective flow-through nanohole array-based biosensing platform for the label-free detection of uropathogenic E. coli in real time. Biosens Bioelectron 2018; 106:105–110 [View Article] [PubMed]
    [Google Scholar]
  152. Naseri M, Halder A, Mohammadniaei M, Prado M, Ashley J et al. A multivalent aptamer-based electrochemical biosensor for biomarker detection in urinary tract infection. Electrochimica Acta 2021; 389:138644 [View Article]
    [Google Scholar]
  153. Tombelli S, Minunni M, Mascini M. Analytical applications of aptamers. Biosens Bioelectron 2005; 20:2424–2434 [View Article] [PubMed]
    [Google Scholar]
  154. Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990; 249:505–510 [View Article] [PubMed]
    [Google Scholar]
  155. Ravindranath SP, Wang Y, Irudayaraj J. SERS driven cross-platform based multiplex pathogen detection. Sens Actuators B Chem 2011; 152:183–190 [View Article]
    [Google Scholar]
  156. Su W, Lin M, Lee H, Cho M, Choe W-S et al. Determination of endotoxin through an aptamer-based impedance biosensor. Biosens Bioelectron 2012; 32:32–36 [View Article] [PubMed]
    [Google Scholar]
  157. Queirós RB, de-los-Santos-Álvarez N, Noronha JP, Sales MGF. A label-free DNA aptamer-based impedance biosensor for the detection of E. coli outer membrane proteins. Sens Actuators B Chem 2013; 181:766–772 [View Article]
    [Google Scholar]
  158. Novoselov KS, Geim AK, Morozov SV, Jiang D, Zhang Y et al. Electric field effect in atomically thin carbon films. Science 2004; 306:666–669 [View Article] [PubMed]
    [Google Scholar]
  159. Zhang X, Jing Q, Ao S, Schneider GF, Kireev D et al. Ultrasensitive field‐effect biosensors enabled by the unique electronic properties of graphene. Small 2020; 16:15 [View Article]
    [Google Scholar]
  160. Ohno Y, Maehashi K, Matsumoto K. Label-free biosensors based on aptamer-modified graphene field-effect transistors. J Am Chem Soc 2010; 132:18012–18013 [View Article] [PubMed]
    [Google Scholar]
  161. Jiang Z, Feng B, Xu J, Qing T, Zhang P et al. Graphene biosensors for bacterial and viral pathogens. Biosens Bioelectron 2020; 166:112471 [View Article] [PubMed]
    [Google Scholar]
  162. Wang Y, Bai X, Wen W, Zhang X, Wang S. Ultrasensitive electrochemical biosensor for HIV gene detection based on graphene stabilized gold nanoclusters with exonuclease amplification. ACS Appl Mater Interfaces 2015; 7:18872–18879 [View Article]
    [Google Scholar]
  163. Schedin F, Geim AK, Morozov SV, Hill EW, Blake P et al. Detection of individual gas molecules adsorbed on graphene. Nat Mater 2007; 6:652–655 [View Article] [PubMed]
    [Google Scholar]
  164. Thakur B, Zhou G, Chang J, Pu H, Jin B et al. Rapid detection of single E. coli bacteria using a graphene-based field-effect transistor device. Biosens Bioelectron 2018; 110:16–22 [View Article] [PubMed]
    [Google Scholar]
  165. Keihan AH, Hosseinzadeh G, Sajjadi S, Ashiani D, Dashtestani F et al. Bacteriophage-based biosensor for detection of E. coli bacteria on graphene modified carbon paste electrode. Nanosci and NanoAsia 2019; 9:408–413 [View Article]
    [Google Scholar]
  166. Farzin L, Sadjadi S, Shamsipur M, Sheibani S. Electrochemical genosensor based on carbon nanotube/amine-ionic liquid functionalized reduced graphene oxide nanoplatform for detection of human papillomavirus (HPV16)-related head and neck cancer. J Pharm Biomed Anal 2020; 179:112989 [View Article] [PubMed]
    [Google Scholar]
  167. Jia F, Duan N, Wu S, Dai R, Wang Z et al. Impedimetric Salmonella aptasensor using a glassy carbon electrode modified with an electrodeposited composite consisting of reduced graphene oxide and carbon nanotubes. Microchim Acta 2016; 183:337–344 [View Article]
    [Google Scholar]
  168. Shahrokhian S, Ranjbar S. Development of a sensitive diagnostic device based on zeolitic imidazolate frameworks-8 using ferrocene–graphene oxide as electroactive indicator for Pseudomonas aeruginosa detection. ACS Sustainable Chem Eng 2019; 7:12760–12769 [View Article]
    [Google Scholar]
  169. Chen Y, Li Y, Yang Y, Wu F, Cao J et al. A polyaniline-reduced graphene oxide nanocomposite as a redox nanoprobe in a voltammetric DNA biosensor for Mycobacterium tuberculosis. Microchim Acta 2017; 184:1801–1808 [View Article]
    [Google Scholar]
  170. Ji J, Pang Y, Li D, Wang X, Xu Y et al. Single-layered graphene/au-nanoparticles-based love wave biosensor for highly sensitive and specific detection of Staphylococcus aureus gene sequences. ACS Appl Mater Interfaces 2020; 12:12417–12425 [View Article] [PubMed]
    [Google Scholar]
  171. Ghorbanpoor H, Dizaji AN, Akcakoca I, Blair EO, Ozturk Y et al. A fully integrated rapid on-chip antibiotic susceptibility test – A case study for Mycobacterium smegmatis. Sens Actuator A Phys 2022; 339:113515 [View Article]
    [Google Scholar]
  172. Labib M, Sargent EH, Kelley SO. Electrochemical methods for the analysis of clinically relevant biomolecules. Chem Rev 2016; 116:9001–9090 [View Article] [PubMed]
    [Google Scholar]
  173. Kim K, Lee S, Ryu C-M. Interspecific bacterial sensing through airborne signals modulates locomotion and drug resistance. Nat Commun 2013; 4: [View Article]
    [Google Scholar]
  174. Aathithan S, Plant JC, Chaudry AN, French GL. Diagnosis of bacteriuria by detection of volatile organic compounds in urine using an automated headspace analyzer with multiple conducting polymer sensors. J Clin Microbiol 2001; 39:2590–2593 [View Article] [PubMed]
    [Google Scholar]
  175. Kodogiannis VS, Lygouras JN, Tarczynski A, Chowdrey HS. Artificial odor discrimination system using electronic nose and neural networks for the identification of urinary tract infection. IEEE Trans Inf Technol Biomed 2008; 12:707–713 [View Article] [PubMed]
    [Google Scholar]
  176. Pavlou AK, Magan N, McNulty C, Jones J, Sharp D et al. Use of an electronic nose system for diagnoses of urinary tract infections. Biosens Bioelectron 2002; 17:893–899 [View Article] [PubMed]
    [Google Scholar]
  177. Roine A, Saviauk T, Kumpulainen P, Karjalainen M, Tuokko A et al. Rapid and accurate detection of urinary pathogens by mobile IMS-based electronic nose: a proof-of-principle study. PLoS One 2014; 9:e114279 [View Article] [PubMed]
    [Google Scholar]
  178. Adebiyi A, Than N, Swaminathan S, Abdi M, Bowers AN et al. Paper presented at the Rapid Strain Differentiation of E. coli-inoculated Urine Using Olfactory-based Smart Sensors. SEIA’2019 Conference Proceedings; 2020: Lulu. com
    [Google Scholar]
  179. Kodogiannis V, Wadge E. The use of gas-sensor arrays to diagnose urinary tract infections. Int J Neural Syst 2005; 15:363–376 [View Article] [PubMed]
    [Google Scholar]
  180. Drabińska N, Hewett K, White P, Avison MB, Persad R et al. Application of a solid-phase microextraction-gas chromatography-mass spectrometry/metal oxide sensor system for detection of antibiotic susceptibility in urinary tract infection-causing Escherichia coli - A proof of principle study. Adv Med Sci 2022; 67:1–9 [View Article] [PubMed]
    [Google Scholar]
  181. Hewett K, Drabińska N, White P, Avison MB, Persad R et al. Towards the identification of antibiotic-resistant bacteria causing urinary tract infections using volatile organic compounds analysis-a pilot study. Antibiotics 2020; 9:797 [View Article] [PubMed]
    [Google Scholar]
  182. Korpi A, Järnberg J, Pasanen A-L. Microbial volatile organic compounds. Crit Rev Toxicol 2009; 39:139–193 [View Article] [PubMed]
    [Google Scholar]
  183. Baev MV, Baev D, Radek AJ, Campbell JW. Growth of Escherichia coli MG1655 on LB medium: monitoring utilization of sugars, alcohols, and organic acids with transcriptional microarrays. Appl Microbiol Biotechnol 2006; 71:310–316 [View Article] [PubMed]
    [Google Scholar]
  184. Lisboa PJ, Taktak AFG. The use of artificial neural networks in decision support in cancer: a systematic review. Neural Netw 2006; 19:408–415 [View Article] [PubMed]
    [Google Scholar]
  185. Saritas I. Prediction of breast cancer using artificial neural networks. J Med Syst 2012; 36:2901–2907 [View Article] [PubMed]
    [Google Scholar]
  186. Saritas I, Ozkan IA, Sert IU. Prognosis of prostate cancer by artificial neural networks. Expert Syst Appl 2010; 37:6646–6650 [View Article]
    [Google Scholar]
  187. Kaya E, Aktan ME, Koru AT, Akdoğan E. Diagnosis of anemia in children via artificial neural network. Int J Intell Syst Appl Eng 2015; 3:24 [View Article]
    [Google Scholar]
  188. Ozkan IA, Koklu M, Sert IU. Diagnosis of urinary tract infection based on artificial intelligence methods. Comput Methods Programs Biomed 2018; 166:51–59 [View Article] [PubMed]
    [Google Scholar]
  189. Lv J, Deng S, Zhang L. A review of artificial intelligence applications for antimicrobial resistance. Biosaf Health 2021; 3:22–31 [View Article]
    [Google Scholar]
  190. Healthy.io Detect UTIs with smartphone-powered clinical urine testing; 2022 https://healthy.io/eu/services/uti/ accessed 22 September 2022
  191. Burke AE, Thaler KM, Geva M, Adiri Y. Feasibility and acceptability of home use of a smartphone-based urine testing application among women in prenatal care. Am J Obstet Gynecol 2019; 221:527–528 [View Article] [PubMed]
    [Google Scholar]
  192. Goździkiewicz N, Zwolińska D, Polak-Jonkisz D. The use of artificial intelligence algorithms in the diagnosis of urinary tract infections-a literature review. J Clin Med 2022; 11:10 [View Article] [PubMed]
    [Google Scholar]
  193. Wand ME, Taylor HV, Auer JL, Bock LJ, Hind CK et al. Evaluating the level of nitroreductase activity in clinical Klebsiella pneumoniae isolates to support strategies for nitro drug and prodrug development. Int J Antimicrob Agents 2019; 54:538–546 [View Article] [PubMed]
    [Google Scholar]
  194. McGeer A, Campbell B, Emori TG, Hierholzer WJ, Jackson MM et al. Definitions of infection for surveillance in long-term care facilities. Am J Infect Control 1991; 19:1–7 [View Article] [PubMed]
    [Google Scholar]
  195. Stone ND, Ashraf MS, Calder J, Crnich CJ, Crossley K et al. Surveillance definitions of infections in long-term care facilities: revisiting the McGeer criteria. Infect Control Hosp Epidemiol 2012; 33:965–977 [View Article]
    [Google Scholar]
  196. Stapleton AE, Stroud MR, Hakomori SI, Stamm WE. The globoseries glycosphingolipid sialosyl galactosyl globoside is found in urinary tract tissues and is a preferred binding receptor In vitro for uropathogenic Escherichia coli expressing pap-encoded adhesins. Infect Immun 1998; 66:3856–3861 [View Article] [PubMed]
    [Google Scholar]
  197. Ofek I, Mirelman D, Sharon N. Adherence of Escherichia coli to human mucosal cells mediated by mannose receptors. Nature 1977; 265:623–625 [View Article] [PubMed]
    [Google Scholar]
  198. Prasadarao NV, Wass CA, Hacker J, Jann K, Kim KS. Adhesion of S-fimbriated Escherichia coli to brain glycolipids mediated by sfaA gene-encoded protein of S-fimbriae. J Biol Chem 1993; 268:10356–10363 [PubMed]
    [Google Scholar]
  199. Khan AS, Kniep B, Oelschlaeger TA, Van Die I, Korhonen T et al. Receptor structure for F1C fimbriae of uropathogenic Escherichia coli. Infect Immun 2000; 68:3541–3547 [View Article] [PubMed]
    [Google Scholar]
  200. Hasan RJ, Pawelczyk E, Urvil PT, Venkatarajan MS, Goluszko P et al. Structure-function analysis of decay-accelerating factor: identification of residues important for binding of the Escherichia coli Dr adhesin and complement regulation. Infect Immun 2002; 70:4485–4493 [View Article] [PubMed]
    [Google Scholar]
  201. Van Dijk WC, Verbrugh HA, van der Tol ME, Peters R, Verhoef J. Role of Escherichia coli K capsular antigens during complement activation, C3 fixation, and opsonization. Infect Immun 1979; 25:603–609 [View Article] [PubMed]
    [Google Scholar]
  202. Carbonetti NH, Boonchai S, Parry SH, Väisänen-Rhen V, Korhonen TK et al. Aerobactin-mediated iron uptake by Escherichia coli isolates from human extraintestinal infections. Infect Immun 1986; 51:966–968 [View Article] [PubMed]
    [Google Scholar]
  203. Jung N, Byun HJ, Park JH, Kim JS, Kim HW et al. Diagnostic accuracy of urinary biomarkers in infants younger than 3 months with urinary tract infection. Korean J Pediatr 2018; 61:24 [View Article]
    [Google Scholar]
  204. Furlani B, Kouter K, Rozman D, Videtič Paska A. Sequencing of nucleic acids: from the first human genome to next generation sequencing in {COVID}-19 pandemic. Acta Chim Slov 2021; 68:268–278 [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jmm/10.1099/jmm.0.001780
Loading
/content/journal/jmm/10.1099/jmm.0.001780
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error