Next Article in Journal
Cell-Free Synthetic Biology Platform for Engineering Synthetic Biological Circuits and Systems
Previous Article in Journal
Electroextraction of Insoluble Proteins from the Organic Matrix of the Nacreous Layer of the Japanese Pearl Oyster, Pinctada fucata
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Protocol

Study Protocol for a Pilot, Open-Label, Prospective, and Observational Study to Evaluate the Pharmacokinetics of Drugs Administered to Patients during Extracorporeal Circulation; Potential of In Vivo Cytochrome P450 Phenotyping to Optimise Pharmacotherapy

1
Centre for Integrated Preclinical Drug Development, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, 4072 Brisbane, Australia
2
Adult Intensive Care Services, The Prince Charles Hospital, 4032 Chermside, Australia
3
Critical Care Research Group, The Prince Charles Hospital, 4032 Chermside, Australia
4
Department of Cardiothoracic Surgery, The Prince Charles Hospital, 4032 Chermside, Australia
5
Department of Anesthesia, The Prince Charles Hospital, 4032 Chermside, Australia
6
School of Pharmacy, Faculty of Health and Behavioral Sciences, The University of Queensland, 4072 Brisbane, Australia
*
Author to whom correspondence should be addressed.
Methods Protoc. 2019, 2(2), 38; https://doi.org/10.3390/mps2020038
Submission received: 25 February 2019 / Revised: 5 May 2019 / Accepted: 7 May 2019 / Published: 13 May 2019

Abstract

:
Pharmacokinetic alterations of medications administered during surgeries involving cardiopulmonary bypass (CPB) and extracorporeal membrane oxygenation (ECMO) have been reported. The impact of CPB on the cytochrome P450 (CYP) enzymes’ activity is the key factor. The metabolic rates of caffeine, dextromethorphan, midazolam, omeprazole, and Losartan to the CYP-specific metabolites are validated measures of in vivo CYP 1A2, 2D6, 3A4, 2C19, and 2C9 activities, respectively. The study aim is to assess the activities of major CYPs in patients on extracorporeal circulation (EC). This is a pilot, prospective, open-label, observational study in patients undergoing surgery using EC and patients undergoing laparoscopic cholecystectomy as a control group. CYP activities will be measured on the day, and 1–2 days pre-surgery/3–4 days post-surgery (cardiac surgery and Laparoscopic cholecystectomy) and 1–2 days after starting ECMO, 1–2 weeks after starting ECMO, and 1–2 days after discontinuation from ECMO. Aforementioned CYP substrates will be administered to the patient and blood samples will be collected at 0, 1, 2, 4, and 6 h post-dose. Major CYP enzymes’ activities will be compared in each participant on the day, and before/after surgery. The CYP activities will be compared in three study groups to investigate the impact of CYPs on EC.

1. Introduction

More than 1 million cardiac operations using CPB are performed annually worldwide [1]. A major benefit of CPB is that it provides a bloodless heart for cardiac surgery, with blood flow diverted to an extracorporeal circuit. While CPB usually lasts a few hours, extracorporeal membrane oxygenation (ECMO), a modified application of CPB concept, provides longer-term extracorporeal life support (days to weeks) as a bridge to recovery or a bridge to transplant or a destination device [1]. Both CPB and ECMO necessitate exteriorisation of significant blood volumes to achieve extracorporeal oxygenation. This process results in significant pathophysiological alterations, not all of which are clearly understood [2].
Both CPB and ECMO may induce postoperative pathophysiological changes related to systemic inflammatory response syndrome (SIRS) and/or a multiple organ dysfunction syndrome (MODS) due to several interlinked mechanisms such as the exposure of blood to non-physiologic surfaces, coagulopathy, surgical trauma, anaesthesia, increased intestinal permeability to endotoxins, ischemia/reperfusion injury, hypotension, capillary leakage, and multiple organ injury [2,3,4,5,6,7]. Khabar et al. [3] discussed the link between the contribution of potential pathological post-CPB phenomena, including endotoxin and the pro-inflammatory cytokines, TNF-α, IL-6, and IL-8 as their plasma concentrations were increased significantly compared with their pre-CPB levels [3]. A significant increase in the ratio of pro-inflammatory cytokines (IL-6, IL-8, and TNF-α) to the major anti-inflammatory cytokine (IL-10) at seven days after CPB compared with a non-CPB control group was reported in another study [8]. The rapid rise in plasma concentrations of TNF-α and IL-8 almost immediately (within 15 min) after the initiation of ECMO suggests that these cytokines may be important mediators in the development of ECMO-related inflammation [2,7]. In an animal model, IL-6 concentrations in the lungs have been shown to increase after Veno-venous (VV) –ECMO [9]. In other work, the concentrations of the pro-inflammatory interleukins (IL), IL-1β and IL-8, increased in broncho alveolar lavage fluid during VA (Veno arterial)-ECMO treatment. In other work, the plasma concentrations of the major anti-inflammatory cytokine, IL-10, were significantly higher in the broncho alveolar lavage fluid of VV-treated animals [10]. There are several examples in the clinical literature whereby cytokines have been implicated in altering cytochrome P450 (CYP) isoenzyme activities during cardiopulmonary bypass [11] resulting in impaired metabolism of clinically important drugs leading to their accumulation in the body and potential the potential for causing adverse effects.
Therefore, pharmacokinetics (PK) and pharmacodynamics (PD) of administered drugs can be significantly affected in setting of an extracorporeal circuit. Multiple studies conducted in patients undergoing CPB have reported on altered drug disposition [12,13,14,15]. During cardiac surgeries using CPB, patients undergo major physiological changes including hypothermia, haemodilution, reduced blood flow and pressure, release of stress-reactant hormones, fluid and electrolyte shifts, as well as release of cytokines and nitric oxide. CPB institution thus has profound effects on the distribution and elimination of drugs, consequently affecting plasma drug concentrations [15]. These effects constantly change during surgeries using CPB and some continue to exert their influence after the patient has been successfully separated from CPB. Drugs that have critical roles in determining patients’ health and surgical outcomes such as anaesthetics, opioid analgesics, neuromuscular blockers, and antibiotics, are reportedly affected during and after surgery using CPB [11].
As with CPB, significant physiological changes occur in patients on ECMO, which may impact drug disposition and the pharmacological effect. ECMO alters PK by increasing the volume of distribution (Vd) through several mechanisms including drug adsorptive losses onto components of the circuit, haemodilution, and other physiological changes, and impaired drug elimination and sequestering drugs in the ECMO circuit [16]. Haemodilution from priming solutions on commencement of ECMO, ongoing blood product transfusions, sequestration of drugs to the circuit, and administration of fluids to maintain circuit flows has a greater effect on Vd of drugs whose distribution is limited to the plasma compartment; this is particularly relevant for hydrophilic drugs [16,17]. ECMO is also associated with reduced drug elimination and a decrease in the clearance of drugs, which is thought to occur by several mechanisms including renal dysfunction in ECMO patients, reduced metabolism, and reduced regional liver blood flow especially for drugs with a high hepatic extraction ratio as reviewed by Shekar et al. in 2012 [17]. Lack of response or toxicity as a result of altered PK of critically important medications during and post-extracorporeal circulation can potentially risk patients’ lives or extend the recovery process.
Although PK alterations on CPB and ECMO have been studied, there are sparse data on metabolism of administered drugs in these patients. It is possible that such profound physiologic insult will affect drug metabolism significantly. Therefore, we hypothesised that CPB and ECMO will significantly alter the activity of CYP enzymes that catalyse drug metabolism in humans. CPB and ECMO will increase pro-inflammatory cytokine concentrations and inhibit CYP activity to alter the pharmacokinetics of important drugs administered to patients in the peri-operative period.
The aim of this pilot clinical study is to explore a possible link between the inflammatory response caused by CPB or ECMO and CYP isoenzyme inhibition leading to changes in the PK of critically important medications. In this study, a well-characterised drug metabolism phenotyping cocktail comprising single low doses of substrates of 5 major CYP isoenzymes (reference to few CYP phenotyping cocktails that used the same probes), will be administered in patients undergoing treatments involving CPB or ECMO. The pilot study will provide preliminary information regarding the extent of changes and their significance in the activities of major CYP isoenzymes in patients undergoing cardiac surgery with CPB, and/or during the ECMO therapy. To gain insight on the impact of surgery (and not cardiopulmonary circulation) on the activities of CYP isoenzymes, patients undergoing heart surgery without CPB or ECMO are required; however, this is not practical in current cardiac practice. Therefore, a different patient population with similar demographic variables as a control group; viz patients undergoing laparoscopic cholecystectomy will be recruited.

2. Experimental Design

Using an in vivo CYP phenotyping procedure, the activities of CYP isoenzymes on the day of surgery will be compared 1–2 days before or 3–4 days after surgery involving CPB. For patients on ECMO, CYP phenotyping will be conducted at 1–2 days and 1–2 weeks after commencement of ECMO and 1–2 days after termination of ECMO therapy. Patients scheduled to undergo laparoscopic cholecystectomy will be recruited as a control group and CYP phenotyping will be conducted on the day of surgery, and 1–2 days before/3–4 days after surgery (Figure 1). The study is a prospective, open-label, observational study that will recruit patients in the CPB group, the ECMO group, and in the control group at the Prince Charles Hospital, Brisbane, QLD, Australia. Inflammatory and pro-inflammatory cytokines including IL-1β, IL-12p70, IL-10, IL-8, IL-6, IL-4, IL-2, TNF-α, IL-13, and INF-γ will be measured in plasma samples collected at time zero of each CYP phenotyping stage.

3. Procedure

3.1. Objectives

  • To describe the activities of CYP enzymes responsible for metabolism of clinically important drugs following CPB or ECMO support.
  • To explore the impact of cytokine release during CPB and ECMO on the changes in the activities of CYP enzymes.

3.2. Participants

Informed consent will be obtained from study participants or surrogate decision makers as applicable. Eligible patients will be eligible for cardiac surgery using CPB, or ICU patients requiring ECMO, or laparoscopic cholecystectomy admitted at the Prince Charles Hospital, Brisbane, QLD, Australia. A small number of participants (20 participants in CPB and control group and 10 in ECMO group) are expected to be recruited in the study. Due to the large number of patients admitted to the hospital for cardiac surgery and laparoscopic cholecystectomy, the target numbers of participants are achievable. The recruitment of patients on ECMO may be slower due to the unpredictable attendance of participants.
  • Inclusion Criteria
    • Age >18 years and <90 years;
    • Eligible for elective mitral valve or aortic root surgeries involving CPB; or
    • Eligible for ECMO support; or
    • Eligible for elective laparoscopic cholecystectomy.
  • Exclusion Criteria
    • No consent;
    • Pregnancy;
    • Serum bilirubin >150 µmol/L;
    • Already enrolled in an interventional ECMO-related research study (as per HREC approval);
    • Ongoing massive blood transfusion requirement (>50% blood volume transfused in the previous 8 h);
    • Therapeutic plasma exchange in the preceding 24 h;
    • Adverse reaction to any elements of the study drug mixture;
    • People with cognitive impairment or mental illness;
    • Patients with significant coronary artery disease and/or aortic stenosis;
    • Patients with liver disease/dysfunction;
    • Using drugs that are known to be strong inhibitors or inducers of CYP enzymes (Flockhart table);
    • Smokers.

3.3. Phenotyping Cocktail Administration

After informed consent is obtained, 1–2 days before/3–4 days after surgery, an indwelling venous catheter (18 G) will be inserted to the participant’s vein (forearm) for blood sampling. One blood sample (5 mL or one teaspoon) will be taken using the same catheter and participants will then be administered the in vivo drug metabolism phenotyping cocktail that will contain the following drugs:
  • Caffeine: 50 mg, Caffeine Liquid 20 mg/mL, 2.5 mL will be administered to the patient;
  • Dextromethorphan: 30 mg, 10 mg/5 mL, 15 mL via oral dosing syringe will be administered to the patient;
  • Midazolam: 1 mg, 5 mg/5 mL, solution for injection: 1 mL in oral dosing syringe will be administered to the patient;
  • Losartan: 5 mg, 50 mg tablet will be crashed using a mortar and pestle. Powder will be dissolved in 10 mL of water; 1 mL (5 mg) via oral dosing syringe will be administered to the patient;
  • Omeprazole, 20 mg, one 20 mg tablet will be administered orally to the patient.
Four extra blood samples (total volume of 14 mL) will be taken from participants at 1, 2, 4, and 6 h after phenotyping cocktail administration using the indwelling venous catheter (Figure 2).
The same procedure (administration of phenotyping cocktail and collection of blood samples) will be repeated on the day of surgery. The control group of patients will receive the phenotyping cocktail and blood sampling will be conducted on two occasions; viz 1–2 days before/3–4 days after, and on the day of laparoscopic cholecystectomy surgery.
For ECMO patients, a peripheral arterial line will already be in place. At 1–2 days after starting ECMO therapy, one blood sample (3.5 mL) will be taken using the existing arterial line followed by the administration of the drug phenotyping cocktail. Four extra blood samplings will be conducted at 1, 2, 4, and 6 h after administration of the phenotyping cocktail. The CYP phenotyping procedure will be repeated at 1–2 weeks after commencement of ECMO therapy, and 1–2 days after termination of ECMO therapy (Figure 2).
Blood samples will be collected in Blue top tube (sodium citrate as the anticoagulant; 3.5 mL) and will be centrifuged at 3000 x g as soon as possible after collection, and plasma will be separated and transferred to pre-labelled eppendorf tubes. Plasma samples will be transferred into a freezer to be stored at –80 °C until the time of analysis. Patients will be closely monitored for unlikely adverse effects by clinical staff. Prescribed medications of the study participants will be unchanged during this pilot study.
Data Collection and Management
An electronic database will be maintained by the study site. The data to be collected are shown in Table 1.
For each patient, various de-identified clinical and demographic data will be collected by trained research staff onto a data collection form (Table 2 and Table 3). The original paper copy of forms will be stored securely at the investigation site. The electronic database will be protected using a password. Only chief investigators will have access to the data.
Sample Analysis
Validated liquid chromatography tandem mass spectrometry (LC-MS/MS) bioanalytical methods, which have been developed previously, will be used to analyse samples [18]. All samples will be assayed alongside calibration standards and quality control samples, and assays will meet the acceptance criteria based on EMA guidelines [19]. All plasma samples will be stored in the investigation site for seven years after the completion of the study.

3.4. Data Analysis

The activities of CYP1A2, CYP2C19, CYP2D6, CYP3A, and CYP2C9 will be assessed as per the metrics outlined below.
  • CYP2D6: Ratio of area under the plasma concentration—time curve of dextrorphan from 0 to 6 h after drug administration (AUC0–6h) to dextromethorphan AUC0–6h.
  • CYP3A: Ratio of 1′-hydroxymidazolam AUC0-6h to midazolam AUC0–6h.
  • CYP2C9: Ratio of E-3174 (losartan carboxy acid) AUC0-6h to losartan AUC0-6h.
  • CYP1A2: Ratio of paraxanthine AUC0-6h to caffeine AUC0–6h (µmol/L).
  • CYP2C19: Ratio of 5-hydroxyomeprazole AUC0-6h to omeprazole AUC0–6h.
Adverse Events
As low single doses of probe drugs are used in most phenotyping cocktails, this ensures patient/subject safety and minimises the potential for probe drug-drug interactions. No side effects have been reported in previous studies that used caffeine, dextromethorphan, omeprazole, midazolam, and losartan as probe drugs in CYP phenotyping cocktails [20,21].
The drugs that will be administered to patient as part of this study are either standard therapeutic doses or low single doses. Serious adverse events are unlikely given the nature of the medications administered and doses used. Serious adverse events will be reported to the reviewing HREC immediately. In addition, a summary of serious adverse events will be submitted to the reviewing HREC every six months.
Statistical Analysis
As the study is of an exploratory and observational design, it may not be possible to conduct full statistical analysis. The CYP activities will be compared in each study group 1–2 days before/3–4 days after surgery and on the day of surgery and also between the CPB and ECMO groups compared with the laparoscopic cholecystectomy control group using non-parametric Mann–Whitney U test.

4. Ethical and Dissemination

Consent from patients or their legally authorized representatives will be obtained by experienced clinicians and all questions and concerns from patients’ family members and their caregivers will be addressed in a timely manner. Drugs that will be administered to participants as the CYP substrates have been in the market for many years and all their possible side effects regarding their multiple dosing administrations are known [20,22,23,24]. The research application was approved by the Prince Charles Hospital, Metro North Hospital and Health Service (HREC/16/QPCH/39) and the University of Queensland (Clearance no. 2016001643) Human Research Ethics Committees. After the study is completed, the database will be closed and followed by statistical analyses, interpretation of results, and dissemination to scientific journals.
Patient and Public Involvement
Neither patients nor public were involved in the development of the research question, study design, outcome measures, recruitment to and conduct of the study, or assessment of the burden of the intervention. The results of the study will be disseminated to the interested study participants by means of lectures given by the investigators.

5. Discussion

This proposed exploratory clinical study will use an in vivo phenotyping drug cocktail approach to provide key data on the extent to which CPB and/or ECMO alter drug metabolising enzyme activities in patients undergoing EC support. Lack of knowledge about how CPB/ECMO impacts the activities of important drug metabolising enzymes may lead to suboptimal pharmacotherapy and may result in therapeutic failure or drug toxicities. Significant changes, if observed in the activities of CYP isoenzymes in this pilot study, may provide justification to run a subsequent larger-cohort study with adequate power to evaluate the extent of changes on CYP activities along with other variables using PK modelling techniques.
The phenotyping cocktail approach is used to assess the effects of drugs on CYP enzyme activity in vivo and it is a validated and well-tolerated method to measure CYP activities in vivo. This phenotyping cocktail approach has been used in clinical studies such as:
  • Assessment of the potential for metabolic drug-drug interactions of novel drugs [25,26];
  • Identification of the main CYPs involved in the metabolism of particular drugs of interest [27,28,29];
  • Assessment of changes in CYP activities in disease states such as congestive heart failure (CHF), human immunodeficiency virus (HIV)-AIDS, and various types of cancer [30,31,32,33];
  • Evaluation of the impact of herbal medicines and aging on major CYP enzyme activities [34,35,36,37];
  • Enhancement of our collective understanding of the impact of drug metabolism on pharmacotherapy in psychiatry [38].
Most drug metabolism phenotyping cocktails used during the last two decades have been shown to be safe due to the low doses used in patients and also the lack of drug–drug interactions between the probe compounds used to phenotype individual CYPs [39]. One exception is the case of a severe adverse reaction in healthy female volunteers after administration of a phenotyping cocktail containing tramadol, omeprazole, losartan, and caffeine as probe drugs for CYP2D6, CYP2C19, CYP2C9, and CYP1A2, respectively [39]. These unexpected adverse reactions were attributed to tramadol and the authors warned against the combination of tramadol with omeprazole, losartan, and caffeine.
The findings from our proposed pilot study, together with the subsequent prospective larger-cohort clinical study, are anticipated to provide the data enabling development of a guideline to optimise pharmacotherapy in patients undergoing extracorporeal circulation. The number of participants to be recruited is based on the expected number of admitted patients in the hospital in the period of study. If we see high variability in the CYP activities that does not provide enough information for the calculation of the power of the subsequent study, we need to extend the study to recruit more patients. The findings from this study will be novel and invaluable in providing evidence-based dosing schedule for drugs administered during treatment involving EC to improve patient outcomes, reduce mortality, length of stay in ICU, and costs associated with the treatment of complications caused by suboptimal drug therapy.

6. Conclusions

Appreciating CYP activity during EC is a key step in understanding altered PK in patients supported with CPB and ECMO life support. The knowledge generated through this pilot study may lead to development of PK models that incorporate these alterations in cytochrome P450 activity, EC life support, and critical illness-induced PK changes to generate robust drug dosing guidelines in patients undergoing cardiac surgery and/or EC life support.

Author Contributions

S.G., K.S. and J.F.F. designed the study. K.S. and J.F.F. are responsible for coordination and supervision of the clinical stage of the study. K.S. was responsible for obtaining informed consents from potential participants or authorised surrogates. S.G. and S.K.S.A. were responsible for coordinating and writing the manuscripts. S.K.S.A. is responsible for collection of samples and patients’ data and analysis of the CYP substrates and metabolites using LC-MS/MS method. M.T.S. is assisting in pharmacokinetic assessment of the study and reviewing the manuscript. R.N., I.R., and P.T. are assisting with the protocol development. S.B. assisted in writing pharmacy manual.

Funding

J.F.F. acknowledges fellowship from the Office of Health and Medical research, Queensland Government, Australia. Therapeutic Innovation Australia (TIA) supported SG financially. The Australian Government through the National Collaborative Research Infrastructure Strategy (NCRIS) program supports TIA. SA received International Postgraduate Research Scholarship (IPRS) and University of Queensland Centennial Scholarship (UQCent).

Acknowledgments

We thank Paul Jarrett, India Lye, Sheena Tom and Amanda Corley for assisting in the patient recruitment and blood collection. We also thank all the patients and their advisers participating in this study. The study was supported by a grant from the Prince Charles Hospital Foundation, Brisbane, Queensland, Australia.

Conflicts of Interest

The authors declare no conflict of interest.

Ethics Approval and Consent to Participate

The Prince Charles Hospital, Metro North Hospital and Health Service (HREC/16/QPCH/39) and the University of Queensland (Clearance no. 2016001643) Human Research Ethics Committees approved the study protocol.

Abbreviations

CPBCardiopulmonary bypass
PKPharmacokinetics
VdVolume of Distribution
SIRSSystemic Inflammatory Response Syndrome
MODLMultiple Organ Dysfunction Syndrome
CYPCytochrome P450
ECMOExtracorporeal membrane oxygenation
TNF-αTumor necrotic factor-alpha
ILInterleukin
VV-ECMOVeno venous extracorporeal membrane oxygenation
VA-ECMOVeno arterial extracorporeal membrane oxygenation
ICUIntensive Care Unit
mgmilli grams
mLmilli litre
APACHEAcute Physiology and Chronic Health Evaluation
SOFASequential Organ Failure Assessment
LC-MS/MSliquid chromatography tandem mass spectrometry
FDAFood and Drugs Administration
EMAEuropean medicines agency
ECextracorporeal circulations

References

  1. Punjabi, P.P.; Taylor, K.M. The Science and Practice of Cardiopulmonary Bypass: From Cross Circulation to Ecmo and Sirs. Glob. Cardiol. Sci. Pract. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  2. Millar, J.E.; Fanning, J.P.; McDonald, C.I.; McAuley, D.F.; Fraser, J.F. The Inflammatory Response to Extracorporeal Membrane Oxygenation (Ecmo): A Review of the Pathophysiology. Crit. Care 2016, 20, 387. [Google Scholar] [CrossRef] [PubMed]
  3. Khabar, K.S.; Elbarbary, M.A.; Khouqeer, F.; Devol, E.; Al-Gain, S.; Al-Halees, Z. Circulating Endotoxin and Cytokines after Cardiopulmonary Bypass: Differential Correlation with Duration of Bypass and Systemic Inflammatory Response/Multiple Organ Dysfunction Syndromes. Clin. Immunol. Immunopathol. 1997, 85, 97–103. [Google Scholar] [CrossRef] [PubMed]
  4. Elahi, M.M.; Khan, J.S.; Matata, B.M. Deleterious Effects of Cardiopulmonary Bypass in Coronary Artery Surgery and Scientific Interpretation of Off-Pump’s Logic. Acute Card. Care 2006, 8, 196–209. [Google Scholar] [CrossRef] [PubMed]
  5. Wan, S.; LeClerc, J.L.; Vincent, J.L. Inflammatory Response to Cardiopulmonary Bypass: Mechanisms Involved and Possible Therapeutic Strategies. Chest 1997, 112, 676–692. [Google Scholar] [CrossRef] [PubMed]
  6. Hirthler, M.; Simoni, J.; Dickson, M. Elevated Levels of Endotoxin, Oxygen-Derived Free Radicals, and Cytokines During Extracorporeal Membrane Oxygenation. J. Pediatr. Surg. 1992, 27, 1199–1202. [Google Scholar] [CrossRef]
  7. McILwain, R.B.; Timpa, J.G.; Kurundkar, A.R.; Holt, D.W.; Kelly, D.R.; Hartman, Y.E.; Neel, M.L.; Karnatak, R.K.; Schelonka, R.L.; Anantharamaiah, G.M.; et al. Plasma Concentrations of Inflammatory Cytokines Rise Rapidly During Ecmo-Related Sirs Due to the Release of Preformed Stores in the Intestine. Lab. Investig. 2010, 90, 128–139. [Google Scholar] [CrossRef]
  8. Gasz, B.; Lenard, L.; Racz, B.; Benko, L.; Borsiczky, B.; Cserepes, B.; Gal, J.; Jancso, G.; Lantos, J.; Ghosh, S.; et al. Effect of Cardiopulmonary Bypass on Cytokine Network and Myocardial Cytokine Production. Clin. Cardiol. 2006, 29, 311–315. [Google Scholar] [CrossRef] [PubMed]
  9. Shi, J.; Chen, Q.; Yu, W.; Shen, J.; Gong, J.; He, C.; Hu, Y.; Zhang, J.; Gao, T.; Xi, F.; et al. Continuous Renal Replacement Therapy Reduces the Systemic and Pulmonary Inflammation Induced by Venovenous Extracorporeal Membrane Oxygenation in a Porcine Model. Artif. Organs 2014, 38, 215–223. [Google Scholar] [CrossRef]
  10. Golej, J.; Winter, P.; Schoffmann, G.; Kahlbacher, H.; Stoll, E.; Boigner, H.; Trittenwein, G. Impact of Extracorporeal Membrane Oxygenation Modality on Cytokine Release During Rescue from Infant Hypoxia. Shock 2003, 20, 110–115. [Google Scholar] [CrossRef] [Green Version]
  11. Adiraju, S.K.S.; Shekar, K.; Fraser, J.F.; Smith, M.T.; Ghassabian, S. Effect of Cardiopulmonary Bypass on Cytochrome P450 Enzyme Activity: Implications for Pharmacotherapy. Drug Metab. Rev. 2018, 50, 109–124. [Google Scholar] [CrossRef] [PubMed]
  12. Buylaert, W.A.; Herregods, L.L.; Mortier, E.P.; Bogaert, M.G. Cardiopulmonary Bypass and the Pharmacokinetics of Drugs. An Update. Clin. Pharmacokinet. 1989, 17, 10–26. [Google Scholar] [CrossRef] [PubMed]
  13. Cotogni, P.; Passera, R.; Barbero, C.; Gariboldi, A.; Moscato, D.; Izzo, G.; Rinaldi, M. Intraoperative Vancomycin Pharmacokinetics in Cardiac Surgery with or without Cardiopulmonary Bypass. Ann. Pharmacother. 2013, 47, 455–463. [Google Scholar] [CrossRef] [PubMed]
  14. Nguyen, M.H.; Eells, S.J.; Tan, J.; Sheth, C.T.; Omari, B.; Flores, M.; Wang, J.; Miller, L.G. Prospective, Open-Label Investigation of the Pharmacokinetics of Daptomycin During Cardiopulmonary Bypass Surgery. Antimicrob. Agents Chemother. 2011, 55, 2499–2505. [Google Scholar] [CrossRef]
  15. Van Saet, A.; de Wildt, S.N.; Knibbe, C.A.; Bogers, A.D.; Stolker, R.J.; Tibboel, D. The Effect of Adult and Pediatric Cardiopulmonary Bypass on Pharmacokinetic and Pharmacodynamic Parameters. Curr. Clin. Pharmacol. 2013, 8, 297–318. [Google Scholar] [CrossRef]
  16. Sherwin, J.; Heath, T.; Watt, K. Pharmacokinetics and Dosing of Anti-Infective Drugs in Patients on Extracorporeal Membrane Oxygenation: A Review of the Current Literature. Clin. Ther. 2016, 38, 1976–1994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Shekar, K.; Fraser, J.F.; Smith, M.T.; Roberts, J.A. Pharmacokinetic Changes in Patients Receiving Extracorporeal Membrane Oxygenation. J. Crit. Care 2012, 27, 741.e9–741.e18. [Google Scholar] [CrossRef]
  18. Sreevatsav Adiraju, S.K.; Shekar, K.; Fraser, J.F.; Smith, M.T.; Ghassabian, S. An Improved Lc-Ms/Ms Method for Simultaneous Evaluation of Cyp2c9, Cyp2c19, Cyp2d6 and Cyp3a4 Activity. Bioanalysis 2018, 10, 1577–1590. [Google Scholar] [CrossRef]
  19. European Medicines Agency. Guidance on Bioanalytical Method Validation (21 July 2011); EMA: Amsterdam, The Netherlands, 2011. [Google Scholar]
  20. Ghassabian, S.; Chetty, M.; Tattam, B.N.; Chem, M.C.; Glen, J.; Rahme, J.; Stankovic, Z.; Ramzan, I.; Murray, M.; McLachlan, A.J. A High-Throughput Assay Using Liquid Chromatography-Tandem Mass Spectrometry for Simultaneous in Vivo Phenotyping of 5 Major Cytochrome P450 Enzymes in Patients. Ther. Drug Monit. 2009, 31, 239–246. [Google Scholar] [CrossRef]
  21. Ghassabian, S.; Chetty, M.; Tattam, B.N.; Glen, J.; Rahme, J.; Stankovic, Z.; Ramzan, I.; Murray, M.; McLachlan, A.J. The Participation of Cytochrome P450 3a4 in Clozapine Biotransformation Is Detected in People with Schizophrenia by High-Throughput in Vivo Phenotyping. J. Clin. Psychopharmacol. 2010, 30, 629–631. [Google Scholar] [CrossRef]
  22. Christensen, M.; Andersson, K.; Dalen, P.; Mirghani, R.A.; Muirhead, G.J.; Nordmark, A.; Tybring, G.; Wahlberg, A.; Yasar, U.; Bertilsson, L. The Karolinska Cocktail for Phenotyping of Five Human Cytochrome P450 Enzymes. Clin. Pharmacol. Ther. 2003, 73, 517–528. [Google Scholar] [CrossRef]
  23. Fuhr, U.; Jetter, A.; Kirchheiner, J. Appropriate Phenotyping Procedures for Drug Metabolizing Enzymes and Transporters in Humans and Their Simultaneous Use in the “Cocktail” Approach. Clin. Pharmacol. Ther. 2007, 81, 270–283. [Google Scholar] [CrossRef] [PubMed]
  24. Donzelli, M.; Derungs, A.; Serratore, M.G.; Noppen, C.; Nezic, L.; Krahenbuhl, S.; Haschke, M. The Basel Cocktail for Simultaneous Phenotyping of Human Cytochrome P450 Isoforms in Plasma, Saliva and Dried Blood Spots. Clin. Pharmacokinet. 2014, 53, 271–282. [Google Scholar] [CrossRef]
  25. Ma, X.; Shah, Y.; Cheung, C.; Guo, G.L.; Feigenbaum, L.; Krausz, K.W.; Idle, J.R.; Gonzalez, F.J. The Pregnane X Receptor Gene-Humanized Mouse: A Model for Investigating Drug-Drug Interactions Mediated by Cytochromes P450 3a. Drug Metab. Dispos. 2007, 35, 194–200. [Google Scholar] [CrossRef]
  26. Tomalik-Scharte, D.; Jetter, A.; Kinzig-Schippers, M.; Skott, A.; Sorgel, F.; Klaassen, T.; Kasel, D.; Harlfinger, S.; Doroshyenko, O.; Frank, D.; et al. Effect of Propiverine on Cytochrome P450 Enzymes: A Cocktail Interaction Study in Healthy Volunteers. Drug Metab. Dispos. 2005, 33, 1859–1866. [Google Scholar] [CrossRef] [PubMed]
  27. Eap, C.B.; Bondolfi, G.; Zullino, D.; Bryois, C.; Fuciec, M.; Savary, L.; Jonzier-Perey, M.; Baumann, P. Pharmacokinetic Drug Interaction Potential of Risperidone with Cytochrome P450 Isozymes as Assessed by the Dextromethorphan, the Caffeine, and the Mephenytoin Test. Ther. Drug Monit. 2001, 23, 228–231. [Google Scholar] [CrossRef] [PubMed]
  28. Kashuba, A.D.M.; Nafziger, A.N.; Kearns, G.L.; Leeder, J.S.; Gotschall, R.; Rocci, M.L.; Kulawy, R.W.; Beck, D.J.; Bertino, J.S. Effect of Fluvoxamine Therapy on the Activities of Cyp1a2, Cyp2d6, and Cyp3a as Determined by Phenotyping. Clin. Pharmacol. Ther. 1998, 64, 257–268. [Google Scholar] [CrossRef]
  29. Jetter, A.; Fatkenheuer, G.; Frank, D.; Klaassen, T.; Seeringer, A.; Doroshyenko, O.; Kirchheiner, J.; Hein, W.; Schomig, E.; Fuhr, U.; et al. Do Activities of Cytochrome P450 (Cyp)3a, Cyp2d6 and P-Glycoprotein Differ between Healthy Volunteers and Hiv-Infected Patients? Antiviral Ther. 2010, 15, 975–983. [Google Scholar] [CrossRef]
  30. Frye, R.F.; Schneider, V.M.; Frye, C.S.; Feldman, A.M. Plasma Levels of Tnf-Alpha and Il-6 Are Inversely Related to Cytochrome P450-Dependent Drug Metabolism in Patients with Congestive Heart Failure. J. Card. Fail. 2002, 8, 315–319. [Google Scholar] [CrossRef]
  31. Johnson, B.M.; Song, I.H.; Adkison, K.K.; Borland, J.; Fang, L.; Lou, Y.; Berrey, M.M.; Nafziger, A.N.; Piscitelli, S.C.; Bertino, J.S., Jr. Evaluation of the Drug Interaction Potential of Aplaviroc, a Novel Human Immunodeficiency Virus Entry Inhibitor, Using a Modified Cooperstown 5 + 1 Cocktail. J. Clin. Pharmacol. 2006, 46, 577–587. [Google Scholar] [CrossRef]
  32. De Wit, D.; Gelderblom, H.; Sparreboom, A.; den Hartigh, J.; den Hollander, M.; Konig-Quartel, J.M.; Hessing, T.; Guchelaar, H.J.; van Erp, N.P. Midazolam as a Phenotyping Probe to Predict Sunitinib Exposure in Patients with Cancer. Cancer Chemother. Pharmacol. 2014, 73, 87–96. [Google Scholar] [CrossRef]
  33. Dees, E.C.; Watkins, P.B. Role of Cytochrome P450 Phenotyping in Cancer Treatment. J. Clin. Oncol. 2005, 23, 1053–1055. [Google Scholar] [CrossRef]
  34. Gurley, B.J.; Swain, A.; Hubbard, M.A.; Williams, D.K.; Barone, G.; Hartsfield, F.; Tong, Y.; Carrier, D.J.; Cheboyina, S.; Battu, S.K. Clinical Assessment of Cyp2d6-Mediated Herb-Drug Interactions in Humans: Effects of Milk Thistle, Black Cohosh, Goldenseal, Kava Kava, St. John’s Wort, and Echinacea. Mol. Nutr. Food Res. 2008, 52, 755–763. [Google Scholar] [CrossRef]
  35. Markowitz, J.S.; Donovan, J.L.; DeVane, C.L.; Taylor, R.M.; Ruan, Y.; Wang, J.S.; Chavin, K.D. Effect of St John’s Wort on Drug Metabolism by Induction of Cytochrome P450 3a4 Enzyme. J. Am. Med. Assoc. 2003, 290, 1500–1504. [Google Scholar] [CrossRef]
  36. Williams, M.L.; Bhargava, P.; Cherrouk, I.; Marshall, J.L.; Flockhart, D.A.; Wainer, I.W. A Discordance of the Cytochrome P450 2c19 Genotype and Phenotype in Patients with Advanced Cancer. Br. J. Clin. Pharmacol. 2000, 49, 485–488. [Google Scholar] [CrossRef]
  37. Zadoyan, G.; Rokitta, D.; Klement, S.; Dienel, A.; Hoerr, R.; Gramatte, T.; Fuhr, U. Effect of Ginkgo Biloba Special Extract Egb 761(R) on Human Cytochrome P450 Activity: A Cocktail Interaction Study in Healthy Volunteers. Eur. J. Clin. Pharmacol. 2012, 68, 553–560. [Google Scholar] [CrossRef]
  38. Hiemke, C.; Shams, M. Phenotyping and Genotyping of Drug Metabolism to Guide Pharmacotherapy in Psychiatry. Curr. Drug Deliv. 2013, 10, 46–53. [Google Scholar] [CrossRef]
  39. Pedersen, R.S.; Damkier, P.; Christensen, M.M.; Brosen, K. A Cytochrome P450 Phenotyping Cocktail Causing Unexpected Adverse Reactions in Female Volunteers. Eur. J. Clin. Pharmacol. 2013, 69, 1997–1999. [Google Scholar] [CrossRef]
Figure 1. Blood Sampling Schedule.
Figure 1. Blood Sampling Schedule.
Mps 02 00038 g001
Figure 2. CYP phenotyping schedule for patients on ECMO and cardiac surgery with CPB.
Figure 2. CYP phenotyping schedule for patients on ECMO and cardiac surgery with CPB.
Mps 02 00038 g002
Table 1. Data collection and management.
Table 1. Data collection and management.
Type of DataDetails Recorded
Demographic Data
  • Age
  • Gender
  • Weight
  • Height
  • Food intake
  • Water intake
Clinical Data
  • Admission diagnosis
  • Illness severity scores [Sequential Organ Failure Assessment (SOFA) on the day of sampling & Acute Physiology and Chronic Health Evaluation (APACHE III) on admission].
  • Use of renal replacement therapy for patents on ECMO
  • Other diseases
  • Other medications
Organ Function Fata
  • Serum bilirubin, total protein and albumin concentrations
  • Serum creatinine concentrations
  • 8-hour urinary creatinine clearance
  • 24 h fluid balance and blood product requirements
ECMO Data
  • Days of therapy
  • ECMO flows during sampling period
  • Type of oxygenator and pump
CPB Data
  • Duration of CPB
  • Type of CPB
  • Type of the hemofilter, blood and effluent pump
Table 2. Demographic and clinical collection form for patients on ECMO.
Table 2. Demographic and clinical collection form for patients on ECMO.
Data Collection Form Extracorporeal Circulation Study (ECMO)Patient No.
Days on ECMO:
Days off ECMO:
Date:
Time:
Food intake:
Water intake:
AgeSex (M/F)Weight (Kg)Height (cm)
ICU Admission DiagnosisAPACHE IIISOFA
Days/hours on ECMO
ECMO flow rate
Type of ECMOVA  Mps 02 00038 i001        Mps 02 00038 i001        Mps 02 00038 i001 VV Other
PumpJosta Rota flow Mps 02 00038 i001
Levotronix Centrimag Mps 02 00038 i001
Cardiohelp Mps 02 00038 i001
Oxygenator Quadrox
Other (please specify) ………
Serum bilirubin (µmol/L)RRT: Yes/No
If yes, please specify mode and flow below:
Midazolam Infusion Bolus (YES/NO)
Midazolam continuous infusion (YES/NO)
Serum Albumin (µmol/L)CVVHCVVHDFSCUF
Serum Creatinine (µmol/L)EDDIHDOTHER
Total Protein (g/L)Effluent Flow Rate (mL/h)
Blood Urea (mmol/L)Blood Flow Rate (mL/h)
8h Creatinine Clearance
Blood Product Transfusion Details24 h Fluid Balance
Table 3. Demographic and clinical data collection form for patients on CPB.
Table 3. Demographic and clinical data collection form for patients on CPB.
Data Collection form
Extracorporeal Circulation Study
(CPB)
Patient No.
Date:
Time:
Food intake:
Water intake:
AgeSex (M/F)Weight (Kg)Height (cm)
ICU Admission Diagnosis
Hours on CPB
Type of CPB
Hypothermia
Blood pump flow rate
Type of membrane
Serum bilirubin (µmol/L)8 h Creatinine Clearance
Serum Creatinine (µmol/L)
Serum Albumin (g/L)
Total Proteins (g/L)
Blood Urea (mmol/L)

Share and Cite

MDPI and ACS Style

Adiraju, S.K.S.; Shekar, K.; Tesar, P.; Naidoo, R.; Rapchuk, I.; Belz, S.; Fraser, J.F.; Smith, M.T.; Ghassabian, S. Study Protocol for a Pilot, Open-Label, Prospective, and Observational Study to Evaluate the Pharmacokinetics of Drugs Administered to Patients during Extracorporeal Circulation; Potential of In Vivo Cytochrome P450 Phenotyping to Optimise Pharmacotherapy. Methods Protoc. 2019, 2, 38. https://doi.org/10.3390/mps2020038

AMA Style

Adiraju SKS, Shekar K, Tesar P, Naidoo R, Rapchuk I, Belz S, Fraser JF, Smith MT, Ghassabian S. Study Protocol for a Pilot, Open-Label, Prospective, and Observational Study to Evaluate the Pharmacokinetics of Drugs Administered to Patients during Extracorporeal Circulation; Potential of In Vivo Cytochrome P450 Phenotyping to Optimise Pharmacotherapy. Methods and Protocols. 2019; 2(2):38. https://doi.org/10.3390/mps2020038

Chicago/Turabian Style

Adiraju, Santosh Kumar Sreevatsav, Kiran Shekar, Peter Tesar, Rishendran Naidoo, Ivan Rapchuk, Stephen Belz, John F Fraser, Maree T Smith, and Sussan Ghassabian. 2019. "Study Protocol for a Pilot, Open-Label, Prospective, and Observational Study to Evaluate the Pharmacokinetics of Drugs Administered to Patients during Extracorporeal Circulation; Potential of In Vivo Cytochrome P450 Phenotyping to Optimise Pharmacotherapy" Methods and Protocols 2, no. 2: 38. https://doi.org/10.3390/mps2020038

Article Metrics

Back to TopTop