Display options
Share it on

Ther Drug Monit. 2022 Feb 01;44(1):32-49. doi: 10.1097/FTD.0000000000000934.

Therapeutic Drug Monitoring of Antibiotic Drugs: The Role of the Clinical Laboratory.

Therapeutic drug monitoring

Maria Shipkova, Hedi Jamoussi

Affiliations

  1. Competence Center for Therapeutic Drug Monitoring, SYNLAB Holding Germany GmbH, SYNLAB MVZ Leinfelden-Echterdingen GmbH, Leinfelden-Echterdingen, Germany.

PMID: 34726200 DOI: 10.1097/FTD.0000000000000934

Abstract

BACKGROUND: Therapeutic drug monitoring (TDM) of anti-infective drugs is an increasingly complex field, given that in addition to the patient and drug as 2 usual determinants, its success is driven by the pathogen. Pharmacodynamics is related both to the patient (toxicity) and bacterium (efficacy or antibiotic susceptibility). The specifics of TDM of antimicrobial drugs stress the need for multidisciplinary knowledge and expertise, as in any other field. The role and the responsibility of the laboratory in this interplay are both central and multifaceted. This narrative review highlights the role of the clinical laboratory in the TDM process.

METHODS: A literature search was conducted in PubMed and Google Scholar, focusing on the past 5 years (studies published since 2016) to limit redundancy with previously published review articles. Furthermore, the references cited in identified publications of interest were screened for additional relevant studies and articles.

RESULTS: The authors addressed microbiological methods to determine antibiotic susceptibility, immunochemical and chromatographic methods to measure drug concentrations (primarily in blood samples), and endogenous clinical laboratory biomarkers to monitor treatment efficacy and toxicity. The advantages and disadvantages of these methods are critically discussed, along with existing gaps and future perspectives on strategies to provide clinicians with as reliable and useful results as possible.

CONCLUSIONS: Although interest in the field has been the driver for certain progress in analytical technology and quality in recent years, laboratory professionals and commercial providers persistently encounter numerous unresolved challenges. The main tasks that need tackling include broadly and continuously available, easily operated, and cost-effective tests that offer short turnaround times, combined with reliable and easy-to-interpret results. Various fields of research are currently addressing these features.

Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.

Conflict of interest statement

The authors declare no conflict of interest.

References

  1. Mabilat C, Gros MF, Nicolau D, et al. Diagnostic and medical needs for therapeutic drug monitoring of antibiotics. Eur J Clin Microbiol Infect Dis. 2020;39:791–797. - PubMed
  2. Roberts JA, Paul SK, Akova M, et al. DALI: defining antibiotic levels in intensive care unit patients: are current β-Lactam antibiotic doses sufficient for critically ill patients? Clin Infect Dis. 2014;58:1072–1083. - PubMed
  3. Blot S, Koulenti D, Akova M, et al. Does contemporary vancomycin dosing achieve therapeutic targets in a heterogeneous clinical cohort of critically ill patients? Data from the multinational DALI study. Crit Care. 2014;18:R99. - PubMed
  4. Abdulla A, Dijkstra A, Hunfeld NGM, et al. Failure of target attainment of beta-lactam antibiotics in critically ill patients and associated risk factors: a two-center prospective study (EXPAT). Crit Care.2020;24:558. - PubMed
  5. Ollivier J, Carrié C, d'Houdain N, et al. Are standard dosing regimens of ceftriaxone adapted for critically ill patients with augmented creatinine clearance? Antimicrob Agents Chemother. 2019;63:e02134-18. - PubMed
  6. Carrié C, Petit L, d'Houdain N, et al. Association between augmented renal clearance, antibiotic exposure and clinical outcome in critically ill septic patients receiving high doses of β-lactams administered by continuous infusion: a prospective observational study. Int J Antimicrob Agents. 2018;51:443–449. - PubMed
  7. Fournier A, Eggimann P, Pantet O, et al. Impact of real-time therapeutic drug monitoring on the prescription of antibiotics in burn patients requiring admission to the Intensive care unit. Antimicrob Agents Chemother. 2018;62:e01818-17. - PubMed
  8. Abdul-Aziz MH, Alffenaar JC, Bassetti M, et al. Antimicrobial therapeutic drug monitoring in critically ill adult patients: a position paper. Intensive Care Med. 2020;46:1127–1153. - PubMed
  9. Darwich AS, Polasek TM, Aronson JK, et al. Model-informed precision dosing: background, requirements, validation, implementation, and forward trajectory of individualizing drug therapy. Annu Rev Pharmacol Toxicol. 2021;61:225–245. - PubMed
  10. Wicha SG, Märtson AG, Nielsen EI, et al. From therapeutic drug monitoring to model-informed precision dosing for antibiotics. Clin Pharmacol Ther. 2021;109:928–941. - PubMed
  11. Martínez-González NA, Keizer E, Plate A, et al. Point-of-care C-reactive protein testing to reduce antibiotic prescribing for respiratory tract infections in primary care: systematic review and meta-analysis of randomised controlled trials. Antibiotics (Basel). 2020;16:610. - PubMed
  12. Leicht HB, Weinig E, Mayer B, et al. Ceftriaxone-induced hemolytic anemia with severe renal failure: a case report and review of literature. BMC Pharmacol Toxicol. 2018;19:67. - PubMed
  13. Lee EY, Caffrey AR. Thrombocytopenia with tedizolid and linezolid. Antimicrob Agents Chemother. 2018;62:e01453-17. - PubMed
  14. Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet. 2013;52:1–8. - PubMed
  15. Self WH, Balk RA, Grijalva CG, et al. Procalcitonin as a marker of etiology in adults hospitalized with community-acquired pneumonia. Clin Infect Dis. 2017;65:183–190. - PubMed
  16. Fontela PS, O'Donnell S, Papenburg J. Can biomarkers improve the rational use of antibiotics? Curr Opin Infect Dis. 2018;31:347–352. - PubMed
  17. Petel D, Winters N, Gore GC, et al. Use of C-reactive protein to tailor antibiotic use: a systematic review and meta-analysis. BMJ Open. 2018;8:e022133. - PubMed
  18. Iankova I, Thompson-Leduc P, Kirson NY, et al. Efficacy and safety of procalcitonin guidance in patients with suspected or confirmed sepsis: a systematic review and meta-analysis. Crit Care Med. 2018;46:691–698. - PubMed
  19. Meier MA, Branche A, Neeser OL, et al. Procalcitonin-guided antibiotic treatment in patients with positive blood cultures: a patient-level meta-analysis of randomized trials. Clin Infect Dis. 2019;69:388–396. - PubMed
  20. Wirz Y, Meier MA, Bouadma L, et al. Effect of procalcitonin-guided antibiotic treatment on clinical outcomes in intensive care unit patients with infection and sepsis patients: a patient-level meta-analysis of randomized trials. Crit Care. 2018;22:191. - PubMed
  21. Heffernan AJ, Denny KJ. Host diagnostic biomarkers of infection in the ICU: where are we and where are we going? Curr Infect Dis Rep. 2021;23:4. - PubMed
  22. Scharf C, Paal M, Schroeder I, et al. Therapeutic drug monitoring of meropenem and piperacillin in critical illness-experience and recommendations from one year in routine clinical practice. Antibiotics (Basel). 2020;9:131. - PubMed
  23. Mitaka C. Clinical laboratory differentiation of infectious versus non-infectious systemic inflammatory response syndrome. Clin Chim Acta. 2005;351:17–29. - PubMed
  24. Williams P, Cotta MO, Roberts JA. Pharmacokinetics/pharmacodynamics of β-Lactams and therapeutic drug monitoring: from theory to practical issues in the intensive care unit. Semin Respir Crit Care Med. 2019;40:476–487. - PubMed
  25. Bragadottir G, Redfors B, Ricksten SE. Assessing glomerular filtration rate (GFR) in critically ill patients with acute kidney injury—true GFR versus urinary creatinine clearance and estimating equations. Crit Care. 2013;17:R108. - PubMed
  26. Casu GS, Hites M, Jacobs F, et al. Can changes in renal function predict variations in β-lactam concentrations in septic patients? Int J Antimicrob Agents. 2013;42:422–428. - PubMed
  27. Sunder S, Jayaraman R, Mahapatra HS, et al. Estimation of renal function in the intensive care unit: the covert concepts brought to light. J Intensive Care. 2014;2:31. - PubMed
  28. Weidhase L, Wellhöfer D, Schulze G, et al. Is interleukin-6 a better predictor of successful antibiotic therapy than procalcitonin and C-reactive protein? A single center study in critically ill adults. BMC Infect Dis. 2019;19:150. - PubMed
  29. Molano Franco D, Arevalo-Rodriguez I, Roqué I Figuls M, et al. Plasma interleukin‐6 concentration for the diagnosis of sepsis in critically ill adults. Cochrane Database Syst Rev. 2019;4:CD011811. - PubMed
  30. Hung SK, Lan HM, Han ST, et al. Current evidence and limitation of biomarkers for detecting sepsis and systemic infection. Biomedicines. 2020;8:494. - PubMed
  31. Hombach M, Ochoa C, Maurer FP, et al. Relative contribution of biological variation and technical variables to zone diameter variations of disc diffusion susceptibility testing. J Antimicrob Chemother. 2016;71:141–151. - PubMed
  32. Mouton JW, Meletiadis J, Voss A, et al. Variation of MIC measurements: the contribution of strain and laboratory variability to measurement precision. J Antimicrob Chemother. 2018;73:2374–2379. - PubMed
  33. Mulroney KT, Hall JM, Huang X, et al. Rapid susceptibility profiling of carbapenem-resistant Klebsiella pneumoniae. Sci Rep. 2017;7:1903. - PubMed
  34. Machen A, Drake T, Wang YF. Same day identification and full panel antimicrobial susceptibility testing of bacteria from positive blood culture bottles made possible by a combined lysis-filtration method with MALDI-TOF VITEK mass spectrometry and the VITEK2 system. PLoS One. 2014;9:e87870. - PubMed
  35. Lagacé-Wiens PR, Adam HJ, Karlowsky JA, et al. Identification of blood culture isolates directly from positive blood cultures by use of matrix-assisted laser desorption ionization-time of flight mass spectrometry and a commercial extraction system: analysis of performance, cost, and turnaround time. J Clin Microbiol. 2012;50:3324–3328. - PubMed
  36. Ferreira L, Sánchez-Juanes F, González-Avila M, 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. - PubMed
  37. Griffin PM, Price GR, Schooneveldt JM, et al. Use of matrix-assisted laser desorption ionization-time of flight mass spectrometry to identify vancomycin-resistant enterococci and investigate the epidemiology of an outbreak. J Clin Microbiol. 2012;50:2918–2931. - PubMed
  38. Nix ID, Idelevich EA, Storck LM, et al. Detection of methicillin resistance in Staphylococcus aureus from agar cultures and directly from positive blood cultures using MALDI-TOF mass spectrometry-based direct-on-target microdroplet growth assay. Front Microbiol. 2020;11:232. - PubMed
  39. Sparbier K, Schubert S, Weller U, et al. Matrix-assisted laser desorption ionization-time of flight mass spectrometry-based functional assay for rapid detection of resistance against β-lactam antibiotics. J Clin Microbiol. 2012;50:927–937. - PubMed
  40. Hrabák J, Studentová V, Walková R, et al. Detection of NDM-1, VIM-1, KPC, OXA-48, and OXA-162 carbapenemases by matrix-assisted laser desorption ionization-time of flight mass spectrometry. J Clin Microbiol. 2012;50:2441–2443. - PubMed
  41. Carvalhaes CG, Cayô R, Visconde MF, et al. Detection of carbapenemase activity directly from blood culture vials using MALDI-TOF MS: a quick answer for the right decision. J Antimicrob Chemother. 2014;69:2132–2136. - PubMed
  42. Chong PM, McCorrister SJ, Unger MS, et al. MALDI-TOF MS detection of carbapenemase activity in clinical isolates of Enterobacteriaceae spp., Pseudomonas aeruginosa, and Acinetobacter baumannii compared against the Carba-NP assay. J Microbiol Methods. 2015;111:21–23. - PubMed
  43. Nagy E, Becker S, Sóki J, et al. Differentiation of division I (cfiA-negative) and division II (cfiA-positive) Bacteroides fragilis strains by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. J Med Microbiol. 2011;60:1584–1590. - PubMed
  44. Fenyvesi VS, Urbán E, Bartha N, et al. Use of MALDI-TOF/MS for routine detection of cfiA gene-positive Bacteroides fragilis strains Int J Antimicrob Agents. 2014;44:474–475. - PubMed
  45. Josten M, Dischinger J, Szekat C, et al. Identification of agr-positive methicillin-resistant Staphylococcus aureus harbouring the class A mec complex by MALDI-TOF mass spectrometry. Int J Antimicrob Agents. 2014;304:1018–1023. - PubMed
  46. Khan ZA, Siddiqui MF, Park S. Current and emerging methods of antibiotic susceptibility testing. Diagnostics (Basel). 2019;9:49. - PubMed
  47. Hombach M, Zbinden R, Böttger EC. Standardisation of disk diffusion results for antibiotic susceptibility testing using the sirscan automated zone reader. BMC Microbiol. 2013;13:225. - PubMed
  48. Chen CH, Lu Y, Sin ML, et al. Antimicrobial susceptibility testing using high surface-to-volume ratio microchannels. Anal Chem. 2010;82:1012–1019. - PubMed
  49. Baltekin Ö, Boucharin A, Tano E, et al. Antibiotic susceptibility testing in less than 30 min using direct single-cell imaging. Proc Natl Acad Sci U S A. 2017;114:9170–9175. - PubMed
  50. Marques SM, Esteves da Silva JC. Firefly bioluminescence: a mechanistic approach of luciferase catalyzed reactions. IUBMB Life. 2009;61:6–17. - PubMed
  51. Dong T, Zhao X. Rapid identification and susceptibility testing of uropathogenic microbes via immunosorbent ATP-bioluminescence assay on a microfluidic simulator for antibiotic therapy. Anal Chem. 2015;87:2410–2418. - PubMed
  52. Ivancic V, Mastali M, Percy N, et al. Rapid antimicrobial susceptibility determination of uropathogens in clinical urine specimens by use of ATP bioluminescence. J Clin Microbiol. 2008;46:1213–1219. - PubMed
  53. Peck Palmer OM, Dasgupta A. A review of the preanalytical errors that impact therapeutic drug monitoring. Ther Drug Monit. 2021; 43:595–608. - PubMed
  54. Traugott KA, Maxwell PR, Green K, et al. Effects of therapeutic drug monitoring criteria in a computerized prescriber-order-entry system on the appropriateness of vancomycin level orders. Am J Health Syst Pharm. 2011;68:347–352. - PubMed
  55. Van Vooren S, Verstraete AG. A sensitive and high-throughput quantitative liquid chromatography high-resolution mass spectrometry method for therapeutic drug monitoring of 10 β-lactam antibiotics, linezolid and two β-lactamase inhibitors in human plasma. Biomed Chromatogr. 2021;35:e5092. - PubMed
  56. Lefeuvre S, Bois-Maublanc J, Hocqueloux L, et al. A simple ultra-high-performance liquid chromatography-high resolution mass spectrometry assay for the simultaneous quantification of 15 antibiotics in plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1065-1066:50–58. - PubMed
  57. Rehm S, Rentsch KM. A 2D HPLC-MS/MS method for several antibiotics in blood plasma, plasma water, and diverse tissue samples. Anal Bioanal Chem. 2020;412:715–725. - PubMed
  58. Decosterd LA, Mercier T, Ternon B, et al. Validation and clinical application of a multiplex high performance liquid chromatography—tandem mass spectrometry assay for the monitoring of plasma concentrations of 12 antibiotics in patients with severe bacterial infections. J Chromatogr B Analyt Technol Biomed Life Sci. 2020;1157:122160. - PubMed
  59. Panda BK, Bargaje M, Sathiyanarayanan L. A simple and reliable analytical method for simultaneous quantification of first line antitubercular drugs in human plasma by LCMS/MS. Anal Methods. 2020;12:3909–3917. - PubMed
  60. Tsai IL, Sun HY, Chen GY, et al. Simultaneous quantification of antimicrobial agents for multidrug-resistant bacterial infections in human plasma by ultra-high-pressure liquid chromatography-tandem mass spectrometry. Talanta. 2013;116:593–603. - PubMed
  61. Baietto L, D'Avolio A, Ariaudo A, et al. Development and validation of a new UPLC-PDA method to quantify linezolid in plasma and in dried plasma spots. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;936:42–47. - PubMed
  62. Abdulla A, Bahmany S, Wijma RA, et al. Simultaneous determination of nine β-lactam antibiotics in human plasma by an ultrafast hydrophilic-interaction chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2017;1060:138–143. - PubMed
  63. Tindula RJ, Ambrose PJ, Harralson AF. Aminoglycoside inactivation by penicillins and cephalosporins and its impact on drug-level monitoring. Drug Intell Clin Pharm. 1983;17:906–908. - PubMed
  64. Pickering LK, Rutherford I. Effect of concentration and time upon inactivation of tobramycin, gentamicin, netilmicin and amikacin by azlocillin, carbenicillin, mecillinam, mezlocillin and piperacillin. J Pharmacol Exp Ther. 1981;217:345–349. - PubMed
  65. Zander J, Maier B, Zoller M, et al. Effects of biobanking conditions on six antibiotic substances in human serum assessed by a novel evaluation protocol. Clin Chem Lab Med. 2016;54:265–274. - PubMed
  66. McConeghy KW, Liao S, Clark D, et al. Variability in telavancin cross-reactivity among vancomycin immunoassays. Antimicrob Agents Chemother. 2014;58:7093–7097. - PubMed
  67. Shipkova M, Petrova DT, Rosler AE, et al. Comparability and imprecision of 8 frequently used commercially available immunoassays for therapeutic drug monitoring. Ther Drug Monit.2014;36:433–441. - PubMed
  68. Li M, Ma L, Chen C, et al. Evaluation of assays to measure aminoglycosides in serum: comparison of accuracy and precision based on external quality assessment. Ther Drug Monit. 2020;42:710–715. - PubMed
  69. Castoldi S, Cozzi V, Baldelli S, et al. Comparison of the ARK immunoassay with high-performance liquid chromatography with ultraviolet detection for therapeutic drug monitoring of linezolid. Ther Drug Monit. 2018;40:140–143. - PubMed
  70. Brozmanová H, Kacířová I, Uřinovská R, et al. New liquid chromatography-tandem mass spectrometry method for routine TDM of vancomycin in patients with both normal and impaired renal functions and comparison with results of polarization fluoroimmunoassay in light of varying creatinine concentrations. Clin Chim Acta. 2017;469:136–143. - PubMed
  71. Hoppentocht M, Akkeman OW, Voerman AJ, et al. Optimisation of the sensitivity of an immunoassay analysis for tobramycin in serum. J Appl Bioanal. 2015; 1:123–127. - PubMed
  72. Stankowicz MS, Ibrahim J, Brown DL. Once-daily aminoglycoside dosing: an update on current literature. Am J Health Syst Pharm. 2015;72:1357–1364. - PubMed
  73. Singer B, Stevens RW, Westley BP, et al. Falsely elevated vancomycin-concentration values from enzyme immunoassay leading to treatment failure. Am J Health Syst Pharm. 2020;77:9–13. - PubMed
  74. LeGatt DF, Blakney GB, Higgins TN, et al. The effect of paraproteins and rheumatoid factor on four commercial immunoassays for vancomycin: implications for laboratorians and other health care professionals. Ther Drug Monit. 2012;34:306–311. - PubMed
  75. Gunther M, Saxinger L, Gray M, et al. Two suspected cases of immunoglobulin-mediated interference causing falsely low vancomycin concentrations with the Beckman PETINIA method. Ann Pharmacother. 2013;47:e19. - PubMed
  76. Florin L, Vantilborgh A, Pauwels S, et al. IgM interference in the Abbott iVanco immunoassay: a case report. Clin Chim Acta. 2015;447:32–33. - PubMed
  77. Tate J, Ward G. Interferences in immunoassay. Clin Biochem Rev. 2004;25:105–120. - PubMed
  78. Dijkstra JA, Voerman AJ, Greijdanus B, et al. Immunoassay analysis of kanamycin in serum using the tobramycin kit. Antimicrob Agents Chemother. 2016;60:4646–4651. - PubMed
  79. Fridlund J, Woksepp H, Schön T. A microbiological method for determining serum levels of broad spectrum β-lactam antibiotics in critically ill patients. J Microbiol Methods. 2016;129:23–27. - PubMed
  80. Toullec L, Dupouey J, Vigne C, et al. Analytical interference during cefepime therapeutic drug monitoring in intensive care patient: about a case report. Therapie. 2017;72:587–592. - PubMed
  81. Rigo-Bonnin R, Ribera A, Arbiol-Roca A, et al. Development and validation of a measurement procedure based on ultra-high performance liquid chromatography-tandem mass spectrometry for simultaneous measurement of β-lactam antibiotic concentration in human plasma. Clin Chim Acta. 2017;468:215–224. - PubMed
  82. Kai M, Tanaka R, Suzuki Y, et al. Simultaneous quantification of plasma levels of 12 antimicrobial agents including carbapenem, anti-methicillin-resistant Staphylococcus aureus agent, quinolone and azole used in intensive care unit using UHPLC-MS/MS method. Clin Biochem. 2021;90:40–49. - PubMed
  83. Shipkova M, Svinarov D. LC–MS/MS as a tool for TDM services: where are we? Clin Biochem. 2016;49:1009–1023. - PubMed
  84. Veringa A, Sturkenboom MGG, Dekkers BGJ, et al. LC-MS/MS for therapeutic drug monitoring of anti-infective drugs. Trends Analyt Chem. 2016;84:34–40. - PubMed
  85. Caro YS, Cámara MS, De Zan MM. A review of bioanalytical methods for the therapeutic drug monitoring of β-lactam antibiotics in critically ill patients: evaluation of the approaches used to develop and validate quality attributes. Talanta. 2020;210:120619. - PubMed
  86. Carlier M, Stove V, Wallis SC, et al. Assays for therapeutic drug monitoring of β-lactam antibiotics: a structured review. Int J Antimicrob Agents. 2015;46:367–375. - PubMed
  87. Popowicz ND, O'Halloran SJ, Fitzgerald D, et al. A rapid, LC-MS/MS assay for quantification of piperacillin and tazobactam in human plasma and pleural fluid; application to a clinical pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci. 2018;1081-1082:58–66. - PubMed
  88. Zander J, Maier B, Zoller M, et al. Quantification of linezolid in serum by LC-MS/MS using semi-automated sample preparation and isotope dilution internal standardization. Clin Chem Lab Med. 2014;52:381–389. - PubMed
  89. Sakurai N, Nakamura Y, Kawaguchi H, et al. Measurement of linezolid and its metabolites PNU-142300 and PNU-142586 in human plasma using ultra-performance liquid chromatography method. Chem Pharm Bull (Tokyo). 2019;67:439–444. - PubMed
  90. Kiang TK, Schmitt V, Ensom MH, et al. Therapeutic drug monitoring in interstitial fluid: a feasibility study using a comprehensive panel of drugs. J Pharm Sci. 2012;101:4642–4652. - PubMed
  91. Kiriazopoulos E, Zaharaki S, Vonaparti A, et al. Quantification of three beta-lactam antibiotics in breast milk and human plasma by hydrophilic interaction liquid chromatography/positive-ion electrospray ionization mass spectrometry. Drug Test Anal. 2017;9:1062–1072. - PubMed
  92. Zhang M, Moore GA, Chin PKL, et al. Simultaneous determination of cefalexin, cefazolin, flucloxacillin, and probenecid by liquid chromatography-tandem mass spectrometry for total and unbound concentrations in human plasma. Ther Drug Monit. 2018;40:682–692. - PubMed
  93. Lee K, Jun SH, Han M, et al. Multiplex assay of second-line anti-tuberculosis drugs in dried blood spots using ultra-performance liquid chromatography-tandem mass spectrometry. Ann Lab Med. 2016;36:489–493. - PubMed
  94. Barco S, Castagnola E, Moscatelli A, et al. Volumetric adsorptive microsampling-liquid chromatography tandem mass spectrometry assay for the simultaneous quantification of four antibiotics in human blood: method development, validation and comparison with dried blood spot. J Pharm Biomed Anal. 2017;145:704–710. - PubMed
  95. Moorthy GS, Vedar C, Zane NR, et al. Development and validation of a volumetric absorptive microsampling- liquid chromatography mass spectrometry method for the analysis of cefepime in human whole blood: application to pediatric pharmacokinetic study. J Pharm Biomed Anal. 2020;179:113002. - PubMed
  96. Attia AK, Al-Ghobashy MA, El-Sayed GM, et al. Voltammetric monitoring of linezolid, meropenem and theophylline in plasma. Anal Biochem. 2018;545:54–64. - PubMed
  97. Carlier M, Athanasopoulos A, Borrey D, et al. Proficiency testing for meropenem and piperacillin therapeutic drug monitoring: preliminary results from the Belgian society on infectiology and clinical microbiology pharmacokinetic-pharmacodynamic working group. Ther Drug Monit. 2018;40:156–158. - PubMed
  98. Wallenburg E, Brüggemann RJ, Asouit K, et al. First international quality control programme for laboratories measuring antimicrobial drugs to support dose individualization in critically ill patients. J Antimicrob Chemother. 2021;76:430–433. - PubMed
  99. Schuster C, Sterz S, Teupser D, et al. Multiplex therapeutic drug monitoring by isotope-dilution HPLC-MS/MS of antibiotics in critical illnesses. J Vis Exp. 2018;58148. - PubMed
  100. Bruch R, Chatelle C, Kling A, et al. Clinical on-site monitoring of ß-lactam antibiotics for a personalized antibiotherapy. Sci Rep. 2017;7:3127. - PubMed
  101. Bian S, Zhu B, Rong G, et al. Towards wearable and implantable continuous drug monitoring: a review. J Pharm Anal. 2021;11:1–14. - PubMed
  102. Garzón V, Bustos RH, Pinacho GD. Personalized medicine for antibiotics: the role of nanobiosensors in therapeutic drug monitoring. J Pers Med. 2020;10:147. - PubMed
  103. Ates HC, Roberts JA, Lipman J, et al. On-site therapeutic drug monitoring. Trends Biotechnol. 2020;38:1262–1277. - PubMed
  104. Dorofaeff T, Bandini RM, Lipman J, et al. Uncertainty in antibiotic dosing in critically ill neonate and pediatric patients: can microsampling provide the answers? Clin Ther. 2016;38:1961–1975. - PubMed
  105. van den Elsen SHJ, Oostenbrink LM, Heysell SK, et al. Systematic review of salivary versus blood concentrations of antituberculosis drugs and their potential for salivary therapeutic drug monitoring. Ther Drug Monit. 2018;40:17–37. - PubMed
  106. van den Elsen SHJ, Akkerman OW, Jongedijk EM, et al. Therapeutic drug monitoring using saliva as matrix: an opportunity for linezolid, but challenge for moxifloxacin. Eur Respir J. 2020;55:1901903. - PubMed
  107. Brasier N, Widmer A, Osthoff M, et al. Non-invasive drug monitoring of β-lactam antibiotics using sweat analysis—a pilot study. Front Med (Lausanne). 2020;7:476. - PubMed
  108. Richter D, Weigand M. β-lactam microneedle array biosensors: a new technology on the horizon. Lancet Digit Health. 2019;1:e320–e321. - PubMed

Publication Types