Next Article in Journal
The Production of Antibiotics Must Be Reoriented: Repositioning Old Narrow-Spectrum Antibiotics, Developing New Microbiome-Sparing Antibiotics
Next Article in Special Issue
Pharmacokinetic/Pharmacodynamic Modeling and Application in Antibacterial and Antifungal Pharmacotherapy: A Narrative Review
Previous Article in Journal
Does a New Antibiotic Scheme Improve the Outcome of Staphylococcus aureus-Caused Acute Prosthetic Joint Infections (PJI) Treated with Debridement, Antibiotics and Implant Retention (DAIR)?
Previous Article in Special Issue
Intermittent Short-Term Infusion vs. Continuous Infusion of Piperacillin: Steady State Concentrations in Porcine Cervical Spine Tissue Evaluated by Microdialysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Protein Binding in Translational Antimicrobial Development-Focus on Interspecies Differences

1
Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
2
Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(7), 923; https://doi.org/10.3390/antibiotics11070923
Submission received: 17 June 2022 / Revised: 1 July 2022 / Accepted: 5 July 2022 / Published: 8 July 2022

Abstract

:
Background/Introduction: Plasma protein binding (PPB) continues to be a key aspect of antibiotic development and clinical use. PPB is essential to understand several properties of drug candidates, including antimicrobial activity, drug-drug interaction, drug clearance, volume of distribution, and therapeutic index. Focus areas of the review: In this review, we discuss the basics of PPB, including the main drug binding proteins i.e., Albumin and α-1-acid glycoprotein (AAG). Furthermore, we present the effects of PPB on the antimicrobial activity of antibiotics and the current role of PPB in in vitro pharmacodynamic (PD) models of antibiotics. Moreover, the effect of PPB on the PK/PD of antibiotics has been discussed in this review. A key aspect of this paper is a concise evaluation of PPB between animal species (dog, rat, mouse, rabbit and monkey) and humans. Our statistical analysis of the data available in the literature suggests a significant difference between antibiotic binding in humans and that of dogs or mice, with the majority of measurements from the pre-clinical species falling within five-fold of the human plasma value. Conversely, no significant difference in binding was found between humans and rats, rabbits, or monkeys. This information may be helpful for drug researchers to select the most relevant animal species in which the metabolism of a compound can be studied for extrapolating the results to humans. Furthermore, state-of-the-art methods for determining PPB such as equilibrium dialysis, ultracentrifugation, microdialysis, gel filtration, chromatographic methods and fluorescence spectroscopy are highlighted with their advantages and disadvantages.

1. Introduction

Plasma protein binding (PPB) is essential for predicting and understanding several important properties of novel antibiotics, including volume of distribution, clearance, therapeutic index and drug-drug interaction [1,2,3]. Plasma is composed of several proteins that act as transporters of exogenous and endogenous molecules throughout the circulatory system and, to a lesser extent, in the interstitial space of tissue. Among these proteins, albumin and α-1-acid glycoprotein (AAG) are responsible for most of the small molecular weight drug molecule binding, whereas globulins and lipoproteins are responsible to a lesser extent [4,5]. Figure 1 shows the interaction of drug molecules with plasma protein albumin. Together, they are believed to have a major impact on the pharmacokinetic profile of exogenous compounds, but also play a role as diagnostic markers, highlighting their potential for clinical applications [6]. Human serum albumin (HSA) is the most abundant protein in human plasma and contributes to the maintenance of pH and osmotic pressure in the bloodstream at concentrations of 500 to 700 μM [5]. HSA carries peptides, drugs, fatty acids, bilirubin and other endogenous compounds [7]. The formation of non-covalent complexes of small molecular weight molecules with HSA may prevent susceptibility to excretory pathways, including renal glomerular filtration and enzymatic reactions in the liver and bloodstream [8]. AAG, a minor acute-phase component of plasma, is primarily synthesized by hepatocytes and functions as a carrier protein mainly for basic and neutral lipophilic endogenous compounds [9].
The pharmacological effect of a certain drug is dependent on both its pharmacokinetic (PK) and pharmacodynamic (PD) properties. The effects of PPB on the PK of antibiotics have been studied extensively [10], but whether PPB also affects PD and antimicrobial activity (by reducing the non-protein-bound fraction) continues to be debated [11]. This may be due to a lack of standardized in vitro PD models to quantify the impact of PPB of antimicrobials [12]. These divergent results may be due to the use of different experimental media with different protein amounts and types [13,14] or the heterogeneous methods (time-kill curves, MIC assays, dynamic models) used to study the effects of PPB in vitro. A prerequisite for all models is the precise measurement of the fraction of free antibiotics in order to correctly evaluate the impact of PPB on the PD of an antibiotic.
Here we provide an overview of the effects of PPB on the antimicrobial activity of antibiotics and the role of PPB in in vitro PD models of antibiotics. By comparing human PPB data for a variety of drugs with PPB data from different animal species, we gain insight into the potential of extrapolating PPB and PK/PD data from animals to humans.

2. Protein Binding: General Principles

Several proteins are involved in the protein binding of antibiotics including plasma proteins, extracellular tissue proteins and/or intracellular tissue proteins. In vivo, drug molecules are either bound to plasma or tissue proteins, or in an unbound/free state. Moreover, PPB properties greatly vary between drugs [15,16]. Since bound drugs are too large to penetrate most biological membranes, only unbound fractions can enter the target tissue and exert the expected pharmacological effect. Hence, PPB significantly influences the PD of drugs. Acidic drugs, including β-lactam antibiotics, fluoroquinolones and nonsteroidal anti-inflammatory drugs (NSAIDs), primarily bind to albumin [7,17]. In contrast, alkaline drugs, such as verapamil, propranolol and lidocaine, bind to the acute phase protein AAG. In addition, lipophilic molecules (i.e., steroid hormones) and some acidic drugs (i.e., phenobarbital) also bind to AAG [18,19]. Other proteins such as corticosteroid binding globulin are essential for binding some specific drugs including glucocorticoids, but do not play an essential role in general drug-protein binding [20]. The PPB properties of drugs are further affected by inflammation. Albumin concentrations decrease during inflammation, whereas AAG levels increase [21]. Finally, protein binding processes are influenced by several factors such as pH, temperature and concentration of the drug, which must be considered when investigating PPB properties in vitro.

3. Effect of Plasma Protein Binding on Antibiotics

3.1. Pharmacokinetics

Only the free/unbound fraction of small molecules diffuses into the extravascular space where it exerts pharmacological activity and may cause side effects [22,23,24]. Therefore, PPB may lead to lower concentrations of free antibiotics in the target tissue, which could reduce antimicrobial activity. This effect is enhanced for certain antibiotics with more pronounced PPB [23]. On the other hand, if poorly water-soluble drugs bind to serum proteins, their tissue distribution may improve due to better solubilization. Similarly, PPB may also enhance antimicrobial efficacy, with plasma proteins acting as drug reservoirs, resulting in prolonged duration of drug concentrations above the bacterial minimum inhibitory concentration (MIC) [23,25,26]. However, the concentrations of unbound drugs may never reach therapeutic levels when their binding to plasma proteins is too high or irreversible. Therefore, the binding properties of drugs must be determined prior to the development process [27]. Some antibiotics exhibit variation in their PPB that may alter certain PK parameters, but rarely impact clinical activity [20]. Changes in PPB due to drug-drug interactions can rapidly affect unbound fractions of drugs and be a source of adverse drug reactions [20]. In summary, although the literature usually describes the total concentration of the drug in plasma, information on the free/unbound fraction is of critical importance in evaluating the antimicrobial activity of a drug [2].

3.2. Pharmacodynamics

The effect of PPB on PD of antibiotics can be assessed in various settings. For example, studies assessing the impact of PPB on the efficacy of an antibiotic against a particular pathogen might reveal variations in the corresponding MIC [23]. Although both bacterial growth and killing are dynamic processes, the MIC is a sometimes-inconsistent threshold value that only partially reflects the activity of an antibiotic [28,29]. In contrast, growth estimation and antibiotic-enhanced time–kill analysis provides a better estimate of antimicrobial activity than MIC [29]. Therefore, time–kill curves are recommended as the most reliable experimental method to evaluate the effects of PPB on antimicrobial activity [24,30]. Methodologically, bacterial growth media are spiked with human serum or protein supplements to account for PPB in these experiments. The actual percentage of human serum must be less than 50%, as serum alone may inhibit bacterial growth [23,25,26], but the maximum serum concentration that can be used should be assessed for the particular bacteria [24,31,32,33,34]. Table 1 provides an overview of the studies that assessed the effect of plasma protein binding on antimicrobial activity.

4. Methodologies for Determining PPB

Equilibrium dialysis, ultracentrifugation, microdialysis and ultrafiltration are the most common methods for separating bound and unbound fractions of a drug [53,54]. Table 2 provides a description of these methods, including their advantages and disadvantages.
Equilibrium dialysis is considered the reference method [53,54,55,56], which consists of two dialysis cells separated by a semipermeable dialysis membrane [57,58] (Figure 2). Each compartment is filled with either buffer or plasma. Subsequently, unbound drugs disseminate from the plasma samples into the protein-free buffer until equilibrium is attained. The free/unbound fraction of the drug is then directly calculated from the buffer solution [55].
Another common method for quantifying the free concentration of a drug in plasma is ultrafiltration [57,58]. An ultrafiltration (UF) system consists of two chambers that are divided by a semi-permeable filter, which has different molecular weight capacities for protein filtration. UF is based on the principle of separation of small volumes of protein-free phase, by exerting a centrifugal force to a solution with both proteins and the substance of interest that is present in the upper compartment of a special UF device [57,58]. After separation by centrifugation, the free drug concentration is determined from the protein-free ultrafiltrate located in the lower compartment of the UF device.
In vivo microdialysis allows estimation of unbound drug fractions in the interstitial fluid of various tissues, and quantification of PPB in blood [59]. Microdialysis is based on the principle of diffusion of molecules along their concentration gradient between two compartments [60]. To determine PPB, a microdialysis probe with a semi-permeable membrane at its tip is inserted into a blood vessel [58] (Figure 3). Afterwards, a dialysate buffer is flushed through the probe and the unbound fraction of a drug in plasma diffuses through the semi-permeable membrane. Larger molecules, including proteins such as albumin, cannot pass through the membrane. The maximum size of proteins that can diffuse depends on the molecular weight limit of the semipermeable membrane [61]. The collected microdialysate samples can then be analyzed [58,62].
Ultracentrifugation is commonly used to estimate the free fraction of lipophilic compounds, by dividing human plasma into three layers by ultracentrifugation, which are then analyzed individually [73] (Figure 4).
In gel filtration (originally called the “zonal” method), after passing through a dextran gel column, the drug-protein solution is divided into two sections, one containing the free drug and the other containing the protein and protein-bound drug [74]. However, this technique may underestimate PPB because the bound drug separates from the drug-protein complex during passage through the column [74].
Several chromatographic methods are available for the separation of different proteins, including high-performance liquid chromatography (HPLC) and gel permeation chromatography (GPC). Chromatographic separation through an HSA-immobilized column allows relative ranking by percent binding. Moreover, equilibrium gel filtration (EGF) with liquid chromatography-tandem mass spectrometry detection has been suggested to be used earlier in the drug discovery process to measure the free concentrations of highly bound drugs without the need for radiolabeled compounds [75,76]. EGF provides an extra option for compounds that are unable to achieve equilibrium, that bind with high affinity to saturable proteins in diluted plasma, or that are unstable in plasma [75,76]. EGF consists of equilibrating a dextran column with a small molecule of interest and then introducing to the column a buffer solution containing a macromolecule capable of binding to the small molecule. Fluorescence-based spectroscopy is another method for determining the binding of small molecules to human proteins. This technique relies on small fluorescent molecules with unique spectroscopic and binding properties. Fluorescence intensity differences between the bound and unbound protein can then be detected assess PPB [77].

5. Assessment of Protein Binding of Antibiotics in the Serum and Plasma across Species

For preclinical findings to be translated into clinical applications, the differences in physiological processes between animals and humans are important and must first be understood [41,78,79]. The quality and accuracy of preclinical estimates depend on the similarities in physiology, drug metabolism, distribution, and absorption between animals and humans [80,81]. Though there have been great advancements in pharmacokinetics in recent years, it still remains a challenge to predict all of the pharmacokinetic parameters of a drug in humans from animal studies. Nonetheless, it is possible to make reasonably good predictions under some well-defined conditions. For instance, the intrinsic absorption of a certain drug through the wall of the gastrointestinal tract might be comparable across species, as the nature of the biomembrane of epithelial cells is similar in mammals [82]. Moreover, the absorption process is an interaction between the drug and the biomembrane that also plays its role. However, there are other factors, including pH-dependent solubility and first-pass metabolism that affect absorption and can lead to differences in absorption in different animal species. One example of relatively successful prediction is the renal excretion of drugs in humans using the glomerular filtration rate ratio between humans and animals. Similarly, if the drug is primarily excreted by the liver and the rate of excretion is limited by blood flow in the liver, the clearance of the drug in humans can be predicted by blood flow in the liver. Nevertheless, biochemical parameters, such as protein binding and drug metabolism, are less predictable, and vary considerably among species [80,81,82]. Rodents are often selected for such studies because they have a short lifespan and therefore a large number of animals can be bred quickly, which in turn allows multiple studies to be conducted. [83]. Body weight and height are usually considered important co-variables for the determination of key PK parameters. [84]. Therefore, small animals can be expected to excrete drugs faster than humans when compared on a weight-normalized basis. Other physiological parameters such as body temperature and serum albumin concentration are comparable in different animals and independent of animal size [85].
Table 3 provides an interspecies comparison of protein binding in serum and plasma in humans and in various animals. We performed a Wilcoxon matched-pairs signed rank test between human and animal PPB data and found that there was a significant difference between antibiotic binding in humans and dogs or mice. In contrast, no significant difference in binding was found between humans and rats, rabbits, or monkeys (Table 3). However, in monkeys, only four pairs were included in the analysis, thus limiting the generalizability of our findings. We further calculated the ratio of antibiotic PPB in humans and various animals (Table 4). The greatest difference was observed in dogs (ratio human vs. dog: 1.96). In general, there is a reasonable positive correlation between the PPBs detected for drug molecules in human plasma and those of rats, rabbits, and monkeys, although slightly stronger binding to human plasma proteins was observed compared with those of preclinical species (Figure 5). Therefore, for screening purposes, data collected in rats may provide a suitable initial surrogate for human protein binding. We believe that PPB should always be measured in species of interest for further drug development, such as for studying pharmacokinetics in preclinical settings, for understanding pharmacodynamics in disease models, or for making dose-human predictions.

6. Conclusions

PPB has received little attention in the discovery and development of antimicrobial agents in recent decades. Equilibrium dialysis and microdialysis are common methods used for protein binding studies. Designing studies to investigate PPB from a PK or PD perspective and generating adequate PPB data in the preclinical setting must be carefully planned, especially when a new drug project enters the investigational phase.
The present review provides information on whether animal models are adequate predictors of human PPB by analyzing data from the literature. Rats, rabbits, and monkeys exhibited similar PPB characteristics to humans, whereas significant differences were found in dogs and mice compared with humans. Depending on the drug and drug delivery platform, species-specific physiological differences may result in incorrect extrapolation of drug PK, safety and efficacy. On the other hand, interspecies differences in protein binding as a result of PK variations could indicate potential variations in drug effects when used in human patients. Therefore, we advocate the standardization of experimental settings to study PPB and its effect on PK/PD of novel antibiotics. In addition, we recognize the relevance of interspecies differences in PPB but appreciate the potential of extrapolating from preclinical pharmacology and safety studies in different species to estimate outcomes in humans.

Author Contributions

Conceptualization, H.A. and M.Z.; methodology, H.A, F.B.; formal analysis, M.Z.; resources, M.Z.; data curation, H.A.; writing—original draft preparation, H.A.; writing—review and editing, H.A., F.B., M.Z.; supervision, M.Z.; project administration, M.Z.; funding acquisition, M.Z. All authors have read and agreed to the published version of the manuscript.

Funding

Financial support was provided by: Marie Skłodowska Innovative Training Network (ITN), Training towards Innovative Personalized Antibiotic Therapy (TIPAT). The project was funded by the European Union’s Horizon 2020 research and innovation programme (grant agreement no. 861323).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Benet, L.Z.; Hoener, B. Changes in Plasma Protein Binding Have Little Clinical Relevance. Clin. Pharmacol. Ther. 2002, 71, 115–121. [Google Scholar] [CrossRef] [PubMed]
  2. Smith, D.A.; Di, L.; Kerns, E.H. The Effect of Plasma Protein Binding on In Vivo Efficacy: Misconceptions in Drug Discovery. Nat. Rev. Drug Discov. 2010, 9, 929–939. [Google Scholar] [CrossRef] [PubMed]
  3. Liu, X.; Wright, M.; Hop, C.E.C.A. Rational Use of Plasma Protein and Tissue Binding Data in Drug Design: Miniperspective. J. Med. Chem. 2014, 57, 8238–8248. [Google Scholar] [CrossRef] [PubMed]
  4. Wasan, K.M.; Cassidy, S.M. Role of Plasma Lipoproteins in Modifying the Biological Activity of Hydrophobic Drugs. J. Pharm. Sci. 1998, 87, 411–424. [Google Scholar] [CrossRef] [PubMed]
  5. Tozer, T.N.; Rowland, M. Introduction to Pharmacokinetics and Pharmacodynamics: The Quantitative Basis of Drug Therapy; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2006; ISBN 978-0-7817-5149-0. [Google Scholar]
  6. Filip, Z.; Jan, K.; Vendula, S.; Jana, K.Z.; Kamil, M.; Kamil, K. Albumin and A1-Acid Glycoprotein: Old Acquaintances. Expert Opin. Drug Metab. Toxicol. 2013, 9, 943–954. [Google Scholar] [CrossRef]
  7. Bertucci, C.; Domenici, E. Reversible and Covalent Binding of Drugs to Human Serum Albumin: Methodological Approaches and Physiological Relevance. Curr. Med. Chem. 2002, 9, 1463–1481. [Google Scholar] [CrossRef]
  8. Fournier, T.; Medjoubi-N, N.; Porquet, D. Alpha-1-Acid Glycoprotein. Biochim. Biophys. Acta 2000, 1482, 157–171. [Google Scholar] [CrossRef]
  9. Kremer, J.M.; Wilting, J.; Janssen, L.H. Drug Binding to Human Alpha-1-Acid Glycoprotein in Health and Disease. Pharmacol. Rev. 1988, 40, 1–47. [Google Scholar]
  10. Wanat, K. Biological Barriers, and the Influence of Protein Binding on the Passage of Drugs across Them. Mol. Biol. Rep. 2020, 47, 3221–3231. [Google Scholar] [CrossRef] [Green Version]
  11. Di, L. An Update on the Importance of Plasma Protein Binding in Drug Discovery and Development. Expert Opin. Drug Discov. 2021, 16, 1453–1465. [Google Scholar] [CrossRef]
  12. Choi, Y.H.; Zhang, C.; Liu, Z.; Tu, M.-J.; Yu, A.-X.; Yu, A.-M. A Novel Integrated Pharmacokinetic-Pharmacodynamic Model to Evaluate Combination Therapy and Determine In Vivo Synergism. J. Pharmacol. Exp. Ther. 2021, 377, 305–315. [Google Scholar] [CrossRef] [PubMed]
  13. Rubinstein, E.; Diamantstein, L.; Yoseph, G.; Gruzman, G.; Rubinovitch, B.; Barzilai, A.; Keller, N. The Effect of Albumin Globulin, Pus and Dead Bacteria in Aerobic and Anaerobic Conditions on the Antibacterial Activity of Moxifloxacin, Trovafloxacin and Ciprofloxacin against Streptococcus Pneumoniae, Staphylococcus Aureus and Escherichia Coli. Clin. Microbiol. Infect. 2000, 6, 678–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zeitlinger, M.; Sauermann, R.; Fille, M.; Hausdorfer, J.; Leitner, I.; Muller, M. Plasma Protein Binding of Fluoroquinolones Affects Antimicrobial Activity. J. Antimicrob. Chemother. 2008, 61, 561–567. [Google Scholar] [CrossRef] [Green Version]
  15. Musteata, F.M. Calculation of Normalized Drug Concentrations in the Presence of Altered Plasma Protein Binding. Clin. Pharmacokinet. 2012, 51, 55–68. [Google Scholar] [CrossRef]
  16. Celestin, M.N.; Musteata, F.M. Impact of Changes in Free Concentrations and Drug-Protein Binding on Drug Dosing Regimens in Special Populations and Disease States. J. Pharm. Sci. 2021, 110, 3331–3344. [Google Scholar] [CrossRef] [PubMed]
  17. Bergogne-Bérézin, E. Clinical Role of Protein Binding of Quinolones. Clin. Pharmacokinet. 2002, 41, 741–750. [Google Scholar] [CrossRef] [PubMed]
  18. Schley, J.; MÜLler-Oerlinghausen, B. Investigation of the Binding of Various Tricyclic Neuroleptics and Antidepressants to A1-Acid Glycoprotein. J. Pharm. Pharmacol. 2011, 38, 102–106. [Google Scholar] [CrossRef] [PubMed]
  19. Kerkay, J.; Westphal, U. Steroid-Protein XIX. Complex Formation between A1-Acid Steroid Hormones. Biochim. Biophys. Acta BBA-Gen. Subj. 1968, 170, 324–333. [Google Scholar] [CrossRef]
  20. Toutain, P.L.; Bousquet-Melou, A. Free Drug Fraction vs Free Drug Concentration: A Matter of Frequent Confusion. J. Vet. Pharmacol. Ther. 2002, 25, 460–463. [Google Scholar] [CrossRef]
  21. Bailey, D.N.; Briggs, J.R. The Binding of Selected Therapeutic Drugs to Human Serum Alpha-1 Acid Glycoprotein and to Human Serum Albumin In Vitro. Ther. Drug Monit. 2004, 26, 40–43. [Google Scholar] [CrossRef]
  22. Scaglione, F.; Demartini, G.; Arcidiacono, M.M.; Dugnani, S.; Fraschini, F. Influence of Protein Binding on the Pharmacodynamics of Ceftazidime or Ceftriaxone against Gram-Positive and Gram-Negative Bacteria in an In Vitro Infection Model. J. Chemother. 1998, 10, 29–34. [Google Scholar] [CrossRef] [PubMed]
  23. Nix, D.E.; Matthias, K.R.; Ferguson, E.C. Effect of Ertapenem Protein Binding on Killing of Bacteria. Antimicrob. Agents Chemother. 2004, 48, 3419–3424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zeitlinger, M.A.; Sauermann, R.; Traunmüller, F.; Georgopoulos, A.; Müller, M.; Joukhadar, C. Impact of Plasma Protein Binding on Antimicrobial Activity Using Time–Killing Curves. J. Antimicrob. Chemother. 2004, 54, 876–880. [Google Scholar] [CrossRef] [PubMed]
  25. Lang, S.D.; Cameron, G.L.; Mullins, P.R. Anomalous Effect of Serum on the Antimicrobial Activity of Cefoperazone. Drugs 1981, 22 (Suppl. S1), 52–59. [Google Scholar] [CrossRef] [PubMed]
  26. Leggett, J.E.; Craig, W.A. Enhancing Effect of Serum Ultrafiltrate on the Activity of Cephalosporins against Gram-Negative Bacilli. Antimicrob. Agents Chemother. 1989, 33, 35–40. [Google Scholar] [CrossRef] [Green Version]
  27. Davis, R.L. Mechanism of Action and Target Identification: A Matter of Timing in Drug Discovery. iScience 2020, 23, 101487. [Google Scholar] [CrossRef]
  28. Mouton, J.W.; Dudley, M.N.; Cars, O.; Derendorf, H.; Drusano, G.L. Standardization of Pharmacokinetic/Pharmacodynamic (PK/PD) Terminology for Anti-Infective Drugs: An Update. J. Antimicrob. Chemother. 2005, 55, 601–607. [Google Scholar] [CrossRef] [Green Version]
  29. Schmidt, S.; Schuck, E.; Kumar, V.; Burkhardt, O.; Derendorf, H. Integration of Pharmacokinetic/Pharmacodynamic Modeling and Simulation in the Development of New Anti-Infective Agents-Minimum Inhibitory Concentration versus Time-Kill Curves. Expert Opin. Drug Discov. 2007, 2, 849–860. [Google Scholar] [CrossRef]
  30. Schuck, E.L.; Derendorf, H. Pharmacokinetic/Pharmacodynamic Evaluation of Anti-Infective Agents. Expert Rev. Anti-Infect. Ther. 2005, 3, 361–373. [Google Scholar] [CrossRef]
  31. Veronese, F.M.; Bevilacqua, R.; Boccù, E.; Benassi, C.A. Drug-Protein Interaction: The Binding of Cephalosporins to Albumins. Il Farm. Ed. Sci. 1977, 32, 303–310. [Google Scholar]
  32. Svensson, C.K.; Woodruff, M.N.; Baxter, J.G.; Lalka, D. Free Drug Concentration Monitoring in Clinical Practice. Rationale and Current Status. Clin. Pharmacokinet. 1986, 11, 450–469. [Google Scholar] [CrossRef] [PubMed]
  33. Riond, J.L.; Riviere, J.E. Doxycycline Binding to Plasma Albumin of Several Species. J. Vet. Pharmacol. Ther. 1989, 12, 253–260. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, B.L.; Sachdeva, M.; Chambers, H.F. Effect of Protein Binding of Daptomycin on MIC and Antibacterial Activity. Antimicrob. Agents Chemother. 1991, 35, 2505–2508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Palmer, S.M.; Kang, S.L.; Cappelletty, D.M.; Rybak, M.J. Bactericidal Killing Activities of Cefepime, Ceftazidime, Cefotaxime, and Ceftriaxone against Staphylococcus Aureus and Beta-Lactamase-Producing Strains of Enterobacter Aerogenes and Klebsiella Pneumoniae in an In Vitro Infection Model. Antimicrob. Agents Chemother. 1995, 39, 1764–1771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Okamoto, M.P.; Nakahiro, R.K.; Chin, A.; Bedikian, A. Cefepime Clinical Pharmacokinetics: Clin. Pharmacokinet. 1993, 25, 88–102. [Google Scholar] [CrossRef] [PubMed]
  37. Lam, Y.W.; Duroux, M.H.; Gambertoglio, J.G.; Barriere, S.L.; Guglielmo, B.J. Effect of Protein Binding on Serum Bactericidal Activities of Ceftazidime and Cefoperazone in Healthy Volunteers. Antimicrob. Agents Chemother. 1988, 32, 298–302. [Google Scholar] [CrossRef] [Green Version]
  38. Esmieu, F.; Guibert, J.; Rosenkilde, H.C.; Ho, I.; Go, A.L. Pharmacokinetics of Cefotaxime in Normal Human Volunteers. J. Antimicrob. Chemother. 1980, 6, 83–92. [Google Scholar] [CrossRef]
  39. Popick, A.C.; Crouthamel, W.G.; Bekersky, I. Plasma Protein Binding of Ceftriaxone. Xenobiotica 1987, 17, 1139–1145. [Google Scholar] [CrossRef]
  40. Bassler, M.; Blaschke, H.; Just, M.; Daschner, F.D. Effect of Ceftriaxone on Pseudomonas aeruginosa and Staphylococcus aureus in Broth, Serum, and in Combination with Human Polymorphonuclear Leukocytes. Chemotherapy 1982, 28, 390–396. [Google Scholar] [CrossRef]
  41. Schmidt, S.; Röck, K.; Sahre, M.; Burkhardt, O.; Brunner, M.; Lobmeyer, M.T.; Derendorf, H. Effect of Protein Binding on the Pharmacological Activity of Highly Bound Antibiotics. Antimicrob. Agents Chemother. 2008, 52, 3994–4000. [Google Scholar] [CrossRef] [Green Version]
  42. Sevillano, D.; Giménez, M.J.; Alou, L.; Aguilar, L.; Cafini, F.; Torrico, M.; González, N.; Echeverría, O.; Coronel, P.; Prieto, J. Effects of Human Albumin and Serum on the In Vitro Bactericidal Activity of Cefditoren against Penicillin-Resistant Streptococcus Pneumoniae. J. Antimicrob. Chemother. 2007, 60, 156–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wellington, K.; Curran, M.P. Cefditoren Pivoxil: A Review of Its Use in the Treatment of Bacterial Infections. Drugs 2004, 64, 2597–2618. [Google Scholar] [CrossRef] [PubMed]
  44. Zhanel, G.G.; Karlowsky, J.A.; Davidson, R.J.; Hoban, D.J. Effect of Pooled Human Cerebrospinal Fluid on the Postantibiotic Effects of Cefotaxime, Ciprofloxacin, and Gentamicin against Escherichia Coli. Antimicrob. Agents Chemother. 1992, 36, 1136–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kunin, C.M. Clinical Pharmacology of the New Penicillins: I. The Importance of Serum Protein Binding in Determining Antimicrobial Activity and Concentration in Serum. Clin. Pharmacol. Ther. 1966, 7, 166–179. [Google Scholar] [CrossRef] [PubMed]
  46. Baker, M.A.; Schneider, E.K.; Huang, J.X.; Cooper, M.A.; Li, J.; Velkov, T. The Plasma Protein Binding Proteome of Ertapenem: A Novel Compound-Centric Proteomic Approach for Elucidating Drug–Plasma Protein Binding Interactions. ACS Chem. Biol. 2016, 11, 3353–3364. [Google Scholar] [CrossRef] [PubMed]
  47. Dalhoff, A. Contribution of Immunocompetence to the Antibacterial Activities of Ciprofloxacin and Moxifloxacin in an In Vitro Pharmacodynamic Model*. Infection 2005, 33, 44–49. [Google Scholar] [CrossRef] [PubMed]
  48. Laue, H.; Valensise, T.; Seguin, A.; Hawser, S.; Lociuro, S.; Islam, K. Effect of Human Plasma on the Antimicrobial Activity of Iclaprim In Vitro. J. Antimicrob. Chemother. 2007, 60, 1388–1390. [Google Scholar] [CrossRef] [PubMed]
  49. Cafini, F.; Aguilar, L.; González, N.; Giménez, M.J.; Torrico, M.; Alou, L.; Sevillano, D.; Vallejo, P.; Prieto, J. In Vitro Effect of the Presence of Human Albumin or Human Serum on the Bactericidal Activity of Daptomycin against Strains with the Main Resistance Phenotypes in Gram-Positives. J. Antimicrob. Chemother. 2007, 59, 1185–1189. [Google Scholar] [CrossRef]
  50. Cha, R. Influence of Protein Binding under Controlled Conditions on the Bactericidal Activity of Daptomycin in an In Vitro Pharmacodynamic Model. J. Antimicrob. Chemother. 2004, 54, 259–262. [Google Scholar] [CrossRef]
  51. Torrico, M.; Giménez, M.J.; González, N.; Alou, L.; Sevillano, D.; Cafini, F.; Prieto, J.; Cleeland, R.; Aguilar, L. Bactericidal Activity of Daptomycin versus Vancomycin in the Presence of Human Albumin against Vancomycin-Susceptible but Tolerant Methicillin-Resistant Staphylococcus Aureus (MRSA) with Daptomycin Minimum Inhibitory Concentrations of 1–2 μg/mL. Int. J. Antimicrob. Agents 2010, 35, 131–137. [Google Scholar] [CrossRef] [Green Version]
  52. Dykhuizen, R.S.; Harvey, G.; Stephenson, N.; Nathwani, D.; Gould, I.M. Protein Binding and Serum Bactericidal Activities of Vancomycin and Teicoplanin. Antimicrob. Agents Chemother. 1995, 39, 1842–1847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Beer, J.; Wagner, C.C.; Zeitlinger, M. Protein Binding of Antimicrobials: Methods for Quantification and for Investigation of Its Impact on Bacterial Killing. AAPS J. 2009, 11, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Buscher, B.; Laakso, S.; Mascher, H.; Pusecker, K.; Doig, M.; Dillen, L.; Wagner-Redeker, W.; Pfeifer, T.; Delrat, P.; Timmerman, P. Bioanalysis for Plasma Protein Binding Studies in Drug Discovery and Drug Development: Views and Recommendations of the European Bioanalysis Forum. Bioanalysis 2014, 6, 673–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Scholtan, W. Methods of determination and theoretical principles of the serum protein binding of drugs (author’s transl). Arzneimittelforschung 1978, 28, 1037–1047. [Google Scholar] [PubMed]
  56. Oravcová, J.; Böhs, B.; Lindner, W. Drug-Protein Binding Sites. New Trends in Analytical and Experimental Methodology. J. Chromatogr. B Biomed. Appl. 1996, 677, 1–28. [Google Scholar] [CrossRef]
  57. Sebille, B. Methods of Drug Protein Binding Determinations. Fundam. Clin. Pharmacol. 1990, 4 (Suppl. S2), 151s–161s. [Google Scholar] [CrossRef]
  58. Wright, J.D.; Boudinot, F.D.; Ujhelyi, M.R. Measurement and Analysis of Unbound Drug Concentrations. Clin. Pharmacokinet. 1996, 30, 445–462. [Google Scholar] [CrossRef] [PubMed]
  59. Tambe, V.; Pande, S.; Gadeval, A.; Rajpoot, K.; Raval, N.; Tekade, R.K. Microdialysis: An Emerging Technique for Pharmacokinetic–Pharmacodynamic Profiling. In Biopharmaceutics and Pharmacokinetics Considerations; Elsevier: Amsterdam, Netherlands, 2021; pp. 601–616. ISBN 978-0-12-814425-1. [Google Scholar]
  60. Joukhadar, C.; Müller, M. Microdialysis: Current Applications in Clinical Pharmacokinetic Studies and Its Potential Role in the Future. Clin. Pharmacokinet. 2005, 44, 895–913. [Google Scholar] [CrossRef]
  61. Stahl, M.; Bouw, R.; Jackson, A.; Pay, V. Human Microdialysis. Curr. Pharm. Biotechnol. 2002, 3, 165–178. [Google Scholar] [CrossRef]
  62. Verbeeck, R.K. Blood Microdialysis in Pharmacokinetic and Drug Metabolism Studies. Adv. Drug Deliv. Rev. 2000, 45, 217–228. [Google Scholar] [CrossRef]
  63. Heinze, A.; Holzgrabe, U. Determination of the Extent of Protein Binding of Antibiotics by Means of an Automated Continuous Ultrafiltration Method. Int. J. Pharm. 2006, 311, 108–112. [Google Scholar] [CrossRef] [PubMed]
  64. Whitlam, J.B.; Brown, K.F. Ultrafiltration in Serum Protein Binding Determinations. J. Pharm. Sci. 1981, 70, 146–150. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, F.; Xue, J.; Shao, J.; Jia, L. Compilation of 222 Drugs’ Plasma Protein Binding Data and Guidance for Study Designs. Drug Discov. Today 2012, 17, 475–485. [Google Scholar] [CrossRef] [PubMed]
  66. Kwon, Y. Protein Binding. In Handbook of Essential Pharmacokinetics, Pharmacodynamics and Drug Metabolism for Industrial Scientists; Kluwer Academic Publishers: Boston, MA, USA, 2002; pp. 105–120. ISBN 978-0-306-46234-4. [Google Scholar]
  67. Kurz, H.; Trunk, H.; Weitz, B. Evaluation of Methods to Determine Protein-Binding of Drugs. Equilibrium Dialysis, Ultrafiltration, Ultracentrifugation, Gel Filtration. Arzneimittelforschung 1977, 27, 1373–1380. [Google Scholar] [PubMed]
  68. Lin, Z.J.; Musiano, D.; Abbot, A.; Shum, L. In Vitro Plasma Protein Binding Determination of Flunarizine Using Equilibrium Dialysis and Liquid Chromatography-Tandem Mass Spectrometry. J. Pharm. Biomed. Anal. 2005, 37, 757–762. [Google Scholar] [CrossRef]
  69. Wan, H.; Rehngren, M. High-Throughput Screening of Protein Binding by Equilibrium Dialysis Combined with Liquid Chromatography and Mass Spectrometry. J. Chromatogr. A 2006, 1102, 125–134. [Google Scholar] [CrossRef]
  70. Zeitlinger, M.A.; Derendorf, H.; Mouton, J.W.; Cars, O.; Craig, W.A.; Andes, D.; Theuretzbacher, U. Protein Binding: Do We Ever Learn? Antimicrob. Agents Chemother. 2011, 55, 3067–3074. [Google Scholar] [CrossRef] [Green Version]
  71. Ojha, S.; Gaikwad, S.T.; Suthar, T.; Gavane, A. Microbial Bioconversion of Poultry Waste: Value Added Products. Indian J. Pure Appl. Biosci. 2020, 8, 165–173. [Google Scholar] [CrossRef]
  72. Hage, D.S. Chromatographic and Electrophoretic Studies of Protein Binding to Chiral Solutes. J. Chromatogr. A 2001, 906, 459–481. [Google Scholar] [CrossRef]
  73. Nakai, D.; Kumamoto, K.; Sakikawa, C.; Kosaka, T.; Tokui, T. Evaluation of the Protein Binding Ratio of Drugs by a Micro-Scale Ultracentrifugation Method. J. Pharm. Sci. 2004, 93, 847–854. [Google Scholar] [CrossRef]
  74. Bohnert, T.; Gan, L.-S. Plasma Protein Binding: From Discovery to Development. J. Pharm. Sci. 2013, 102, 2953–2994. [Google Scholar] [CrossRef] [PubMed]
  75. Isbell, J.; Yuan, D.; Torrao, L.; Gatlik, E.; Hoffmann, L.; Wipfli, P. Plasma Protein Binding of Highly Bound Drugs Determined With Equilibrium Gel Filtration of Nonradiolabeled Compounds and LC-MS/MS Detection. J. Pharm. Sci. 2019, 108, 1053–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Jadhav, S.; Kumbhar, S.; Bhatia, M. Gel Permeation Chromatographic Method for Drug Protein Binding Studies. Marmara Pharm. J. 2017, 21, 938–948. [Google Scholar] [CrossRef] [Green Version]
  77. McCallum, M.M.; Pawlak, A.J.; Shadrick, W.R.; Simeonov, A.; Jadhav, A.; Yasgar, A.; Maloney, D.J.; Arnold, L.A. A Fluorescence-Based High Throughput Assay for the Determination of Small Molecule−human Serum Albumin Protein Binding. Anal. Bioanal. Chem. 2014, 406, 1867–1875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Bailey, E.M.; Rybak, M.J.; Kaatz, G.W. Comparative Effect of Protein Binding on the Killing Activities of Teicoplanin and Vancomycin. Antimicrob. Agents Chemother. 1991, 35, 1089–1092. [Google Scholar] [CrossRef] [Green Version]
  79. Schmidt, S.; Gonzalez, D.; Derendorf, H. Significance of Protein Binding in Pharmacokinetics and Pharmacodynamics. J. Pharm. Sci. 2010, 99, 1107–1122. [Google Scholar] [CrossRef]
  80. Martignoni, M.; Groothuis, G.M.M.; de Kanter, R. Species Differences between Mouse, Rat, Dog, Monkey and Human CYP-Mediated Drug Metabolism, Inhibition and Induction. Expert Opin. Drug Metab. Toxicol. 2006, 2, 875–894. [Google Scholar] [CrossRef]
  81. Martinez, M.N. Factors Influencing the Use and Interpretation of Animal Models in the Development of Parenteral Drug Delivery Systems. AAPS J. 2011, 13, 632–649. [Google Scholar] [CrossRef] [Green Version]
  82. Lin, J.H. Species Similarities and Differences in Pharmacokinetics. Drug Metab. Dispos. 1995, 23, 1008–1021. [Google Scholar]
  83. Casal, M.; Haskins, M. Large Animal Models and Gene Therapy. Eur. J. Hum. Genet. EJHG 2006, 14, 266–272. [Google Scholar] [CrossRef]
  84. Poggesi, I. Predicting Human Pharmacokinetics from Preclinical Data. Curr. Opin. Drug Discov. Dev. 2004, 7, 100–111. [Google Scholar]
  85. Davies, B.; Morris, T. Physiological Parameters in Laboratory Animals and Humans. Pharm. Res. 1993, 10, 1093–1095. [Google Scholar] [CrossRef] [PubMed]
  86. Walton, K.; Dorne, J.L.C.M.; Renwick, A.G. Species-Specific Uncertainty Factors for Compounds Eliminated Principally by Renal Excretion in Humans. Food Chem. Toxicol. 2004, 42, 261–274. [Google Scholar] [CrossRef] [PubMed]
  87. Ohshima, T.; Hasegawa, T.; Johno, I.; Kitazawa, S. Protein Binding of Cefpiramide in the Plasma of Various Species. J. Pharm. Pharmacol. 1991, 43, 805–806. [Google Scholar] [CrossRef] [PubMed]
  88. Sakamoto, H.; Hatano, K.; Higashi, Y.; Mine, Y.; Nakamoto, S.; Tawara, S.; Kamimura, T.; Matsumoto, F.; Kuwahara, S. Animal Pharmacokinetics of FK037, a Novel Parenteral Broad-Spectrum Cephalosporin. J. Antibiot. 1993, 46, 120–130. [Google Scholar] [CrossRef] [PubMed]
  89. Kita, Y.; Yamazaki, T.; Imada, A. Comparative Pharmacokinetics of SCE-2787 and Related Antibiotics in Experimental Animals. Antimicrob. Agents Chemother. 1992, 36, 2481–2486. [Google Scholar] [CrossRef] [Green Version]
  90. Kita, Y.; Fugono, T.; Imada, A. Comparative Pharmacokinetics of Carumonam and Aztreonam in Mice, Rats, Rabbits, Dogs, and Cynomolgus Monkeys. Antimicrob. Agents Chemother. 1986, 29, 127–134. [Google Scholar] [CrossRef] [Green Version]
  91. Wittendorf, R.W.; Swagzdis, J.E.; Gifford, R.; Mico, B.A. Protein Binding of Glycopeptide Antibiotics with Diverse Physical-Chemical Properties in Mouse, Rat, and Human Serum. J. Pharmacokinet. Biopharm. 1987, 15, 5–13. [Google Scholar] [CrossRef]
  92. Arhin, F.F.; Belley, A.; McKay, G.; Beaulieu, S.; Sarmiento, I.; Parr, T.R.; Moeck, G. Assessment of Oritavancin Serum Protein Binding across Species. Antimicrob. Agents Chemother. 2010, 54, 3481–3483. [Google Scholar] [CrossRef] [Green Version]
  93. Zhanel, G.G.; Kirkpatrick, I.D.; Hoban, D.J.; Kabani, A.M.; Karlowsky, J.A. Influence of Human Serum on Pharmacodynamic Properties of an Investigational Glycopeptide, LY333328, and Comparator Agents against Staphylococcus Aureus. Antimicrob. Agents Chemother. 1998, 42, 2427–2430. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Interaction of drug molecules with plasma protein albumin.
Figure 1. Interaction of drug molecules with plasma protein albumin.
Antibiotics 11 00923 g001
Figure 2. Overview and principle of equilibrium dialysis.
Figure 2. Overview and principle of equilibrium dialysis.
Antibiotics 11 00923 g002
Figure 3. Illustration of the principle of in vitro microdialysis.
Figure 3. Illustration of the principle of in vitro microdialysis.
Antibiotics 11 00923 g003
Figure 4. Schematic illustration of ultracentrifugation.
Figure 4. Schematic illustration of ultracentrifugation.
Antibiotics 11 00923 g004
Figure 5. Scatter plot depicting the correlation between % plasma protein binding in humans and other species. r is the Pearson correlation coefficient.
Figure 5. Scatter plot depicting the correlation between % plasma protein binding in humans and other species. r is the Pearson correlation coefficient.
Antibiotics 11 00923 g005
Table 1. Examples for studies that assessed the effect of plasma protein binding on antimicrobial activity.
Table 1. Examples for studies that assessed the effect of plasma protein binding on antimicrobial activity.
Antibiotic ClassAntibioticPlasma Protein Binding % (Drug Concentration)Protein PreparationMediumTechniques for Protein QuantificationSusceptibility Test/sPathogensReferences
CephalosporinCefepime Ceftazidime Cefotaxime Ceftriaxone19%
21%
38%
84–96% (70–300 µg/mL)
HA (4 g/dL)SMHBNATime–kill curves, MIC, MBECE. aerogenes
K. pneumoniae
S. aureus
[35,36,37,38,39]
Ceftriaxone84–96% (70–300 µg/mL)20% Human serumBHINATime–kill curvesS. aureus
P. aeruginosa
[38,40]
Ceftriaxone ErtapenemHA 76.8 ± 11.0%; BSA, 20.2 ± 8.3%; HSA 56.9 ± 16.6%, HA 73.8 ± 11.6%; BSA 12.4 ± 4.8%; HSA 17.8 ± 11.5%BSA and HSA (40 g/L)MHB, THB + 5% CO2in vitro microdialysisMIC, time–kill curveE. coli
S. pneumoniae
[41]
Cefditoren88%90% HS, 4 g/dL HAMHBNATime–kill curveS. pneumoniae[42,43]
Cefotaxime10–40% (0.5–32 µg/mL)90% Pooled human CSFCAMHBNAMICE. coli[38,44]
Ceftriaxone, Cefoperazone, Moxalactam, Ceftizoxime92.2%
89.7%
63.8%
29.4%
HA (0, 2.5, or 5% solution), heat-inactivated HA (0, 25, 50, or 95%)
(95% Human Serum)
MHBEquilibrium dialysisMIC, MBECS. aureus
E. coli
P. aeruginosa
K. pneumoniae
[26]
PenicillinAmpicillin, Oxacillin20%
60–94%
40 g/L human albuminMHBNATime–kill curveS. aureus[24,45]
CarbapenemErtapenem85–95%
(50–300 µg/mL)
50% pooled human plasmaTSB. CAMHBSurface Plasmon Resonance (SPR) assayMICS. aureus[46]
FluoroquinolonesCiprofloxacin20–40% (2 µg/mL)90% pooled human CSFCAMHBNAMICE. coli[17,44]
Moxifloxacin, Trovafloxacin38% (0.2–5 µg/mL)
77% (0.2–5 µg/mL)
HS (20%, 70%, 100%), HA (4%, 8%, 12%, 16%)MHBUltrafiltrationMICS. aureus
P. aeruginosa
[14]
Ciprofloxacin, Moxifloxacin20–40% (2 µg/mL)
26–30% (1–5 µg/mL)
rat polyvinyl sponge modelBHI,
RPMI 1640 (for cells)
NAViable cell countP. aeruginosa
S. pneumoniae
[14,17,47]
Moxifloxacin
Ciprofloxacin
Trovafloxacin
20–40% (2 µg/mL)
26–30% (1–5 µg/mL)
77% (0.2–5 µg/mL)
HA 10%, 30%, 50%MHBNAMICS. pneumoniae
S. aureus
E. coli
[13,14,17]
DiaminopyrimidineIclaprim93%50% HPMHBNAMICS. aureus[48]
Cyclic lipopeptideDaptomycin91.7%90% HS
4 g/dL HA
MHBNATime–kill curvesS. pneumoniae
E. faecium
[34,49]
Daptomycin91.7%50% HSMHBNAMIC, time–kill curveS. aureus
E. faecium
[34,50]
Daptomycin91.7%4 g/dL HACAMHBNATime–kill curvesMRSA[34,51]
GlycopeptideVancomycin36.9%4 g/dL HACation-adjusted MHBNATime–kill curvesMRSA[51,52]
NA: Not Available; BHI: Brain Heart Infusion; MRSA: Methicillin-resistant Staphylococcus aureus; HA: Human albumin; BSA: Bovine Serum Albumin; HSA: Human Serum Albumin; MHB: Mueller–Hinton Broth; THB: Todd Hewitt broth; MIC: Minimal Inhibitory Concentration; TSB: Tryptone Soy Broth; CAMHB: Cation-Adjusted Mueller–Hinton Broth; HPLC: High-performance liquid chromatography; MBEC: Minimal Biofilm Eradication Concentration; RPMI: Roswell Park Memorial Institute (Growth Media); HP: Haptoglo.
Table 2. Comparison of methods for plasma protein binding (PPB).
Table 2. Comparison of methods for plasma protein binding (PPB).
Technique/MethodPrincipleAdvantagesDrawbacks/IssuesReferences
UltrafiltrationPlasma water and unbound drug is forced through a semipermeable filter, retaining protein–drug complexes
  • Technically simple to perform
  • Requires a small amount of sample
  • Available commercially via kits
  • Quick
  • Does not require the use of unphysiological buffer
  • Inexpensive
  • Suitable for unstable drugs
  • Most commonly used
  • Ultrafiltrate volumes should be ≤40% of the initial plasma sample because of changes in protein concentration
  • Leakage of membrane may happen
  • Membrane adsorption of drugs
  • Partly uncontrolled temperature
[57,58,63,64,65,66]
Equilibrium dialysisSeparated by a semipermeable membrane, unbound drug diffuses from plasma into protein-free buffer, until equilibrium is reached
  • Gold standard method
  • Simple
  • Reliable results
  • Temperature controlled and thermodynamically sound
  • Requires the use of unphysiological buffer
  • Non-specific binding to dialysis device and membrane
  • Volume shifts and pH changes
  • Membrane adsorption
  • Requires time to reach equilibrium
  • Drug stability concerns
[56,58,60,65,66,67,68,69]
MicrodialysisDialysate buffer is driven through an embedded probe, having a microdialysis membrane. Unbound drug disperses from blood into dialysate
  • In vivo protein binding determination
  • Volume shifts or dilution effects are lacking
  • May also be used to find free tissue levels
  • Probe implantation and fixation
  • Altering drug concentration over a period of time
  • Membrane adsorption
  • Needs equilibration in vivo
  • For measurement of PPB of total antibiotic concentration in vivo is needed
[41,56,58,61,62]
UltracentrifugationDissociation of protein and low-molecular-weight components occurs only by gravitation (centrifugation)
  • Simple
  • Lack of membrane adsorption, dilution, volume shifts, drug–protein leakage
  • Does not require use of unphysiological values
  • Buffer
  • Long time span for sample preparation (overnight)
  • Concentration gradient from bottom to top can result in false high binding
  • Less suitable for high molecular weight substances
  • Sedimentation, back diffusion
  • Expensive equipment
[55,56,65,67,70]
Gel filtrationThe free and not the total drug concentration is the independent variable.
  • Robust and high resolution technique
  • Desalination
  • Costly
  • Time-consuming
[71]
Chromatographic methods Chromatographic methods include a range of techniques, based on separation of substances (including the bound and unbound fraction of an antibiotic) on the basis of different physical or chemical properties such as molecular size, charge, affinity etc.
  • Relatively low sample consumption
  • Accurate methods
  • Absence of membrane adsorption, dilution, volume shifts, drug–protein leakage
  • Expensive and elaborate
  • Poorly sensitive for drugs with low affinity binding
[72]
Fluorescence spectroscopyHigher energy photons are used to excite a sample, which then emit lower energy photons. The change in fluorescence at changing ligand/protein concentrations is used to calculate the concentration of bound drug.
  • Enables direct determination of bound drug concentrations
  • Poorly sensitive for drugs with low-affinity binding
  • Elaborate technique
[56]
Table 3. Interspecies comparison of protein binding in serum and plasma. Wilcoxon matched-pairs signed rank test was performed between antibiotics percentage unbound in human and other species. p-Value < 0.05 is considered as significant.
Table 3. Interspecies comparison of protein binding in serum and plasma. Wilcoxon matched-pairs signed rank test was performed between antibiotics percentage unbound in human and other species. p-Value < 0.05 is considered as significant.
For Serum (% Bound)
AntibioticConcentration (µg/mL)HumanDogRatMouseRabbitMonkeyReferences
Cefotetan # 91.03930 [86]
Cefpirmide #1598.44087.4 92.9 [87]
Ceftriaxone #10092.72079.5 93.7 [39]
Cefpirome305.86.815.811.610.3 [88]
Ceftazidime3011.99.517.611.917.6 [88]
Cefzopran208.110.46.47.19.810.9[89]
Cefclidin208.58.510.49.87.38.1[89]
Carumonam20281136202124[90]
Ristocetin4–12073.1 56.153.5 [91]
Oritavancin 82 82.485.3 [92]
Oritavancin # 87.5 80 [93]
Vancomycin4–10034.6 23.417.3 [91]
Mannosylaglycone6–17072.8 87.988.3 [91]
Ardacinaglycone6–18091.5 96.792.1 [91]
Ardacin A6–19097 97.294.6 [91]
Ardacin B6–21098.7 98.597.4 [91]
Ardacin C6–21099.6 99.599.1 [91]
Pseudoaglycone6–19099.4 99.199 [91]
Aztreonam20622085464553[90]
Wilcoxon matched-pairs signed rank test
Number of pairs with human values 9191584
p-value *
(0.0195)
NS
(0.8059)
*
(0.0353)
NS
(0.3750)
NS
(0.5000)
# Plasma was used instead of serum. * p-value < 0.05.
Table 4. Human/animal ratio of protein binding of different antibiotics in serum and plasma. Means difference analysis was performed between antibiotics percentage bound ratio in human and other species. Mean, standard deviation and range of human/animal ratio is provided.
Table 4. Human/animal ratio of protein binding of different antibiotics in serum and plasma. Means difference analysis was performed between antibiotics percentage bound ratio in human and other species. Mean, standard deviation and range of human/animal ratio is provided.
For Serum (% Binding Ratio)
AntibioticHuman/Dog Human/RatHuman/Mouse Human/RabbitHuman/Monkey
Cefotetan #2.33.0
Cefpiramide #2.51.2 1.1
Ceftriaxone #4.61.2 1.0
Cefpirome0.80.40.50.6
Ceftazidime1.20.71.00.7
Cefzopran0.81.31.10.80.7
Cefclidin1.00.80.91.21.0
Carumonam2.50.81.41.31.2
Ristocetin 1.31.4
Oritavancin 1.01.0
Oritavancin # 1.1
Vancomycin 1.52.0
Mannosylaglycone 0.80.8
Ardacinaglycone 0.91.0
Ardacin A 1.01.0
Ardacin B 1.01.0
Ardacin C 1.01.0
Pseudoaglycone 1.01.0
Aztreonam3.10.71.31.31.2
Means difference analysis
Number of pairs with human values9191584
Mean1.961.091.0911.02
Standard deviation1.300.530.330.270.24
Range0.8–4.60.4–3.00.5–2.00.6–1.30.7–1.2
# Plasma was used instead of serum.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ahmed, H.; Bergmann, F.; Zeitlinger, M. Protein Binding in Translational Antimicrobial Development-Focus on Interspecies Differences. Antibiotics 2022, 11, 923. https://doi.org/10.3390/antibiotics11070923

AMA Style

Ahmed H, Bergmann F, Zeitlinger M. Protein Binding in Translational Antimicrobial Development-Focus on Interspecies Differences. Antibiotics. 2022; 11(7):923. https://doi.org/10.3390/antibiotics11070923

Chicago/Turabian Style

Ahmed, Hifza, Felix Bergmann, and Markus Zeitlinger. 2022. "Protein Binding in Translational Antimicrobial Development-Focus on Interspecies Differences" Antibiotics 11, no. 7: 923. https://doi.org/10.3390/antibiotics11070923

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop