Cremophor EL

Cremophor EL Alters the Plasma Protein Binding and Pharmacokinetic Profile of Valspodar in Rats

Author
Ziyad Binkhathlan

ABstrAct Affiliation
Thieme

Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia

Key words
drug delivery, p-glycoprotein, pharmacokinetics
received 22.02.2017 accepted 12.05.2017

Bibliography
DOI https://doi.org/10.1055/s-0043-111411 Published online: 2017
Drug Res
© Georg Thieme Verlag KG Stuttgart · New York ISSN 2194-9379

Correspondence
Dr. Ziyad Binkhathlan King Saud University
Department of Pharmaceutics College of Pharmacy
P. O. Box 2457 11451, Riyadh Saudi Arabia
Tel.: + 966/11/4675 613, Fax: + 966/11/4676 295 [email protected], [email protected]
Cremophor EL is a nonionic surfactant widely used in pharma- ceutical formulations. Nonetheless, there are several reports on the influence of this excipient on the protein binding, phar- macokinetics, and pharmacodynamics of drugs. Valspodar is an investigational non-immunosuppressive derivative of cyclo- sporine A, used in clinical trials for treatment of multidrug re- sistant tumors. The formulation of valspodar (Amdray®) con- tains cremophor EL and ethanol as solubilizing agents. The main aim of the current study was to assess the plasma protein binding (in vitro) and the pharmacokinetic profile of valspodar in the cremophor EL-based formulation in comparison to a cre- mophor EL-free formulation following intravenous (i. v.) admin- istration to rats. Valspodar dissolved in PEG 400/ethanol (di- luted in Dextrose 5 %) was used as the cremophor EL-free formulation. The in vitro plasma unbound fraction (fu) of vals- podar in the cremophor EL formulation was 2.3-fold higher than the PEG 400/ethanol formulation. Following a single i. v. dose of 5 mg/kg, valspodar in the cremophor EL-based formu- lation had around 50 % lower plasma AUC compared to the PEG 400/ethanol formulation. Moreover, the cremophor EL formu- lation had significantly higher volume of distribution and clear- ance in comparison to the PEG 400-based formulation. The results highlight the significance of excipient-drug interaction that should not be overlooked during the early stages of drug development.

Introduction
Cremophor EL is a nonionic surfactant that is commonly used in the pharmaceutical formulations to solubilize hydrophobic drugs in aqueous solutions. Chemically it is produced by reacting castor oil with ethylene oxide at a molar ratio of 1:35 (▶ Fig. 1). Cremo- phor EL is a pale yellow oily liquid with a molecular weight around 3 000 Da, and a hydrophilic-lipophilic balance (HLB) in the range of 12 and 14. Upon addition to an aqueous solution, cremophor EL forms micelles at concentrations above the critical micelle concen- tration (CMC) (~ 90 μg/mL) [1]. Cremophor EL is a major compo- nent in several parenteral pharmaceutical products including Taxol®, Valstar®, Vumon®, Sandimmune®, and Ixempra®.
Cremophor EL is not an inert excipient because it has been shown to inhibit P-glycoprotein (P-gp) in vitro and to increase the AUC of P-gp substrates when given intravenously or orally [2–4] Moreover, cremophor EL has been reported to cause several adverse effects

including hypersensitivity reactions, abnormal lipoprotein patterns, aggregation of erythrocytes and peripheral neuropathy [5].
Valspodar (▶ Fig. 1) is a noncompetitive inhibitor of P-gp, which has been shown to overcome multi-drug resistance (MDR) to vari- ous anticancer agents in vitro and in preclinical and clinical studies [6, 7]. It is a non-immunosuppressive analog of CyA [8–13]. Vals- podar is practically insoluble in water, and also has poor solubility in different organic solvents. Like CyA, the formulation of valspo- dar used in clinic (Amdray®) is composed of cremophor EL and eth- anol. To date, it is not known whether or not cremophor EL, present in the formulation, would have any impact on the pharmacokinet- ic profile of the drug. Therefore, the aim of the current study was to evaluate the plasma protein binding and pharmacokinetic pro- file of valspodar in the cremophor EL/ethanol formulation in com- parison to a cremophor EL-free formulation following intravenous (i. v.) administration to rats.

▶ Fig. 1 Chemical structures of a cremophor EL and b valspodar.

to standard rodent chow and water. All rats were anesthetized using

Materials and Methods
Materials
Valspodar was kindly provided by Novartis (Basel, Switzerland). Cre- mophor EL and PEG 400 were purchased from Sigma-Aldrich (St. Louis, MO, USA). Sodium chloride injection (USP) 0.9 % and hepa- rin sodium for injection (1 000 IU/mL) were obtained from Hospira Inc. (Lake Forest, IL, USA). Acetonitrile, ammonium hydroxide, methanol, and water were all HPLC grade and were purchased from BDH Chemical Ltd (Poole, England). All other chemicals were rea- gent grade.

Methods
Preparation of valspodar formulations
Valspodar formulation was prepared by adding 50 mg of the drug to 600 mg of cremophor EL, and the volume of the mixture was made to 1 mL with ethanol [14]. The formulation was diluted in sa- line to a final concentration of 5 mg/mL before dosing. Cremophor EL-free formulation was prepared by dissolving 50 mg of valspodar in a mixture of PEG 400 and ethanol (2:1), which was then diluted in dextrose 5 % to a final concentration of 5 mg/mL before dosing.

Animals and pharmacokinetic study
The protocols of the animal studies were approved by the Experi- mental Animal Care Centre Review Board (No. C.P.R- 4525) at the College of Pharmacy, King Saud University. Male Sprague-Dawley rats (250–350 g) were kept in cages in temperature-controlled rooms with 12 h light/dark cycles. Animals were given free access
isoflurane and underwent right jugular vein cannulation as previ- ously described [15]. After performing the procedure, the rats were returned to their cages and allowed free access to water, but no food was allowed until 4 h post dose. On the next day, rats were transferred to metabolic cages and divided into 2 groups (3 rats/
group).
Animals were administered valspodar as a single i. v. dose of 5 mg/kg either in its standard cremophor EL/ethanol formulation or in the PEG 400/ethanol formulation. The i. v. dose was injected over 2 min through the cannula, which was immediately followed by an injection of normal saline solution. The first 200 µL volume of blood for the first sample was discarded. The animals were pro- vided free access to food 4 h post dose. Serial blood samples (150– 250 µL) were collected at 0.08, 0.25, 0.75, 1, 2, 4, 6, 9, 12, and 24 h after dosing. Following each sample collection of blood, the can- nula was flushed with heparin diluted in normal saline. Plasma was separated from each blood sample through immediate centrifuga- tion for 3 min; and then stored at -20 °C until analysis. Valspodar plasma concentrations were determined using a liquid chromatog- raphy-mass spectrometry (LC-MS) assay.

Determination of valspodar unbound fraction in vitro Plasma protein binding of valspodar was determined in vitro using the erythrocyte vs. buffer or plasma partitioning method [16, 17]. The blood was obtained from Sprague-Dawley rat. For each formu- lation, the final concentration of valspodar in the samples was 2.5 µg/mL, which is within the range of plasma concentrations ob- tained following administration of 5 mg/kg valspodar to rats [18].

Determination of valspodar concentrations
The concentrations of valspodar in all samples, following liquid-liq- uid extraction, were analyzed by a validated LC-MS assay [18]. The LC-MS system consists of a mass spectrometer (Waters Micromass ZQTM 4000) connected to an HPLC module (Waters 2795) with an autosampler (Milford, Ma, USA). The separation was achieved using a C8 3.5 μm (2.1 × 50 mm) column (Agilent® Eclipse XDB-C8, USA). Mobile phase was composed of a mixture of acetonitrile: ammoni- um hydroxide 0.2 % at a ratio of 90:10 v/v, respectively. The assay lower limit of quantification was 10 ng/mL [18].
For quantitation of valspodar in blood, plasma, and buffer sam- ples, a previously reported liquid-liquid extraction method was used. Briefly, to each 100 µL plasma sample in a glass tube, 500 µL HPLC water, 100 µL sodium hydroxide (1 M), and 50 µL of internal standard (0.25 µg/mL) were added. Diethyl ether/methanol (95:5)

solution (4 mL) was used for extraction of the drug and the internal standard. After vortex-mixing for 30 s followed by a centrifugation at 3 000 × g for 5 min, the organic layer was transferred to clean glass tubes and evaporated using a vacuum concentrator (ISS 110 Speedvac system, Thermosavant). Methanol (250 µL) was used for reconstitution of the residues. The concentration range used for calibration samples was of 0.01–5 µg/mL.

Data and statistical analysis
Standard non-compartmental method was used for determination of the pharmacokinetic parameters of valspodar [17, 19]. Briefly, the elimination rate constant (λz) was estimated by linear regres- sion of the plasma concentrations in the log-linear terminal phase and the corresponding half-life (t1/2) was calculated by dividing 0.693 by λz. The AUC0-∞ was calculated using the combined log-lin- ear trapezoidal rule from time 0 h postdose to the time of the last measured concentration, plus the quotient of the last measured concentration divided by λz. The concentration at time 0 h after i. v. dosing (Cp0) was estimated by extrapolation of the log-linear re- gression line using the first 3 measured plasma concentrations to time 0. The mean residence time (MRT) was calculated by dividing area under the first moment curve (AUMC0-∞) by AUC0-∞, clearance (CL) by dividing dose by AUC0-∞, and volume of distribution at steady-state (Vdss) by multiplying CL by MRT.
The plasma unbound fraction (fu) was determined by using the method illustrated by Schuhmacher and others [16]. Unless other- wise indicated, all data are reported as mean ± SD. Student’s un- paired t-test was used to compare the differences between the means (assuming unequal variance). The level of significance was set at α = 0.05.

Results and Discussion
Valspodar has been widely used in preclinical and clinical studies along with anticancer drugs, that are P-gp substrate, to overcome MDR in tumors. Although the drug has shown success in MDR tu- mor-bearing animal models and has been shown to prolong their survival rate, it was associated with a limited success in clinical tri- als. The formulation of valspodar used in those trials (Amdray®) is composed of cremophor EL and ethanol. The main aim of this study was to evaluate the pharmacokinetic profile of valspodar in the cre- mophor EL-based formulation in comparison to a cremophor EL-
▶ Fig. 2 Plasma concentration vs. time profile in rat following a single iv dose (5 mg/kg( of valspodar in the cremophor EL/Ethanol formulation and PEG 400/Ethanol formulation. Each data point represents the mean ± SD.

free formulation (PEG 400/ethanol) following i. v. administration in rats. Following an i.v. bolus dose of vaslpodar (5 mg/kg) to rats, the pharmacokinetic profile in the cremophor EL/ethanol formula- tion shows a steep decline in plasma concentrations within the first 2 h (▶ Fig. 2). This likely represents a distribution phase, as previ- ous studies showed that valspodar is mainly eliminated by hepatic metabolism and biliary excretion (low extraction ratio), and that only less than 0.1 % of the dose is excreted unchanged in urine [20]. The distribution phase was followed by an elimination phase with a mean t1/2 of approximately 10.55 h. The profile of valspodar in the PEG 400/ethanol formulation showed a less steep decline in plas- ma concentrations at the early time points (up to ~2 h) with a ter- minal phase t1/2 of nearly 10 h (▶ Fig. 2). The difference in the ter- minal phase t1/2 between the two formulations was not statistical- ly significant. Nonetheless, the drug in the cremophor EL-based formulation yielded lower plasma AUC in comparison to the PEG 400/ethanol formulation (▶ table 1).
The cremophor EL/ethanol formulation showed a significantly higher in vitro fu for valspodar (13.66 %) compared to the PEG 400/
ethanol formulation (6.00 %). This increase in fu is likely the cause of the significant increase in the drug’s Vdss (4.73 vs. 2.20 L/kg). And since valspodar is a low extraction ratio drug (~0.27 in rats [19]) and mainly eliminated through hepatic metabolism [20], the increase in its fu resulted in a significant increase in its plasma CL (0.441 vs. 0.225 L/h/kg). The higher plasma CL of valspodar in the cremophor EL-based formulation led to a significantly lower AUC of drug in plasma compared to the PEG 400/ethanol formulation (11.64 vs. 22.97 mg.h/L). Moreover, the elimination t1/2 of valspo- dar was essentially the same for both formulations since the chang- es in Vdss and CL were of the same magnitude (▶ table 1).
Although valspodar is structurally similar to CyA, it has a distinct pharmacokinetic and pharmacodynamic profile compared to CyA. Unlike CyA, valspodar association to erythrocytes was found to be low with a binding coefficient of 1.5, while it was around 61 in the case of CyA [21]. This indicates a low affinity of valspodar to cyclo- philin present in the erythrocytes. This was in line with the human data, where it showed that valspodar binding to human cyclophi-

lin A was at least 100-fold less in comparison to CyA [6]. Further- more, valspodar binding to human cyclophilin B and C was found to be 65- to 85-fold less compared to CyA. And that is likely the rea- son for the lack of immunosuppression and nephrotoxicity with the use of valspodar in clinic [6]. It seems that even cremophor EL af- fects the pharmacokinetics of CyA and valspodar differently. While it is reported that cremophor EL decreases the Vdss and CL of CyA because it decreases the free fraction of drug in blood [22], the op- posite was observed here with valspodar.
It was proposed that presence of cremophor EL, at concentra- tion higher than its critical micelle concentration in blood, keeps CyA entrapped inside the micelles and prevents it from binding to the erythrocytes [22]. This explains the decrease in blood to plas- ma concentration ratios associated with cremophor EL administra- tion to rats. This phenomenon was also observed with paclitaxel i. e., a reduction in fu and blood to plasma concentration ratio [23– 26]. On the other hand, valspodar in the cremophor EL/ethanol for- mulation had lower binding to plasma proteins in vitro, and the higher fu was associated with enhanced partitioning into blood cells as compared to PEG 400/ethanol formulation. The calculated eryth- rocyte to plasma partition coefficients for valspodar in the cremo- phor EL-based formulation and the PEG 400/ethanol formulation were 1.03 ± 0.17 and 0.28 ± 0.11, respectively (▶ table 2). Moreo- ver, the drug’s erythrocytes to buffer partition coefficients were 7.74 ± 1.54 and 5.07 ± 1.63 in the presence and absence of cremo- phor EL, respectively.
The apparent increase in the erythrocyte partitioning in the plasma-free samples has been observed previously with other drugs [26, 27], including valspodar [28]. This is evident by the find- ing that in the presence of plasma proteins, erythrocyte partition- ing of valspodar was markedly impaired regardless of the formula- tion being used (▶ table 2). And that the higher erythrocyte parti- tioning observed with the cremophor EL/ethanol formulation is likely because of the less drug bound to plasma proteins.
We have previously shown that loading valspodar into block co- polymer micelles resulted in a significant reduction in fu compared

to the cremophor EL-based formulation (5.59 vs. 14.85 %, respec- tively) [19]. Nonetheless, the blood to plasma ratio of valspodar in rats was similar in both formulations (~ 0.5), which is close to the fraction of plasma water [19]. Therefore, it seems that cremophor EL does not alter the blood to plasma ratio of valspodar. Moreover, Simon et al. [28] have shown that plasma lipoproteins are the main blood carriers of valspodar and that fluctuations in lipoprotein lev- els would likely result in variations in the drug’s fu and blood distri- bution. And since cremophor EL is known to alter the plasma lipo- proteins [29–32], the plausible reason for the higher fu obtained for valspodar in the cremophor EL/ethanol formulation is that it in- teracts/interferes with the plasma lipoproteins rendering them less available for binding with the drug.
More recently, Liu et al. [33] studied the effect of cremophor EL on the pharmacokinetic properties of seven investigational drugs following i. v. administration in mice and rats. The cremophor EL formulations of the test compounds were prepared differently with a concentration of cremophor EL ranging from 5–20 %. The com- position of the non-cremophor EL formulations were also different, where the test compounds were first solubilized in a co-solvent ei- ther N-Methyl-2-pyrrolidone or PEG 300, or in the complexing agent captisol, and then diluted in 5 % dextrose in water or buffer [33]. The major findings in Liu et al. study was that cremophor EL formulations caused a lower Vdss and CL to different extents in all test compounds, and that these effects are more pronounced in the plasma rather than in blood [33]. And that is because cremo- phor EL micelles likely entrapped the drug and kept it in plasma. Al- though their findings are different than the ones presented in the current study, both studies came to the same conclusion that using cremophor EL as a solubilizer in drug formulations may alter the in- trinsic pharmacokinetic properties of the drug. Furthermore, over- looking such effects at early stages of drug development might have a negative impact on the drug being investigated. And, there- fore, the use of cremophor EL as a drug solubilizer is not recom- mended for i. v. formulation in the early stages of drug develop- ment.

▶ table 1 Plasma pharmacokinetic parameters (mean ± SD) of valspodar in
rats following a single i. v. administration (5 mg/kg). Conclusions

Parameter cremophor EL/
Ethanol PEG 400/Ethanol
AUC0-24h (mg · h/L) 9.93 ± 1.56 20.39 ± 4.98 *
AUC0-∞ (mg · h/L) 11.64 ± 2.39 22.97 ± 5.14 *
t1/2 (h) 10.55 ± 2.08 9.98 ± 1.39
MRT (h) 10.99 ± 2.30 9.73 ± 0.66
CL (L/kg/h) 0.441 ± 0.08 0.225 ± 0.05 *
Vdss (L/kg) 4.73 ± 0.61 2.20 ± 0.58 *
* Denotes significan t difference between groups
Selection of the right excipients in a drug formulation is a crucial step in drug development. Some of the widely used excipients are not inert, and have been reported to alter the pharmacokinetic, pharmacodynamics, and side effect profiles of drugs. In this study, cremophor EL was shown to significantly alter the plasma protein binding and pharmacokinetic profile of valspodar following an i. v. bolus dose in comparison to PEG 400-based formulation. It also showed a significantly higher plasma fu, Vdss, and CL of valspodar as compared to PEG 400/ethanol formulation. These results, as well as the ones previously reported, highlight the significance of excip-

▶ table 2 Unbound fraction (fu) of valspodar in rat plasma in vitro.
Formulation cE/cP cE * /cB fu ( %)
Cremophor EL/Ethanol 1.03 ± 0.17 7.74 ± 1.54 13.66 ± 2.91
PEG 400/Ethanol 0.28 ± 0.11 * 5.07 ± 1.63 * 6.00 ± 2.83 *
* Denotes significant difference between groups; CE/CP: partition coefficients for erythrocyte:plasma; CE * /CB: partition coefficients for erythrocyte:buffer; Data represented as mean ± SD (n = 4)

ient-drug interaction that should not be overlooked during the early stages of drug development. Specifically, the use of cremo- phor EL in i. v. drug formulations is not recommended.

Acknowledgments
The author is grateful to the College of Pharmacy Research Centre and the Deanship of Scientific Research at King Saud University, Ri- yadh, Saudi Arabia for logistic and financial support.

Conflicts of interest

No conflicts of interest to declare.

References

[1]Kessel D. Properties of cremophor EL micelles probed by fluorescence. Photochem Photobiol 1992; 56: 447–451
[2]Woodcock DM, Jefferson S, Linsenmeyer ME et al. Reversal of the multidrug resistance phenotype with cremophor EL, a common vehicle for water-insoluble vitamins and drugs. Cancer Res 1990; 50: 4199–4203
[3]Friche E, Jensen PB, Sehested M et al. The solvents cremophor EL and Tween 80 modulate daunorubicin resistance in the multidrug resistant Ehrlich ascites tumor. Cancer Commun 1990; 2: 297–303
[4]Martin-Facklam M, Burhenne J, Ding R et al. Dose-dependent increase of saquinavir bioavailability by the pharmaceutic aid cremophor EL. Br J Clin Pharmacol 2002; 53: 576–581
[5]Gelderblom H, Verweij J, Nooter K et al. Cremophor EL: the drawbacks and advantages of vehicle selection for drug formulation. Eur J Cancer 2001; 37: 1590–1598
[6]Loor F. Valspodar: current status and perspectives. Expert Opin Investig Drugs 1999; 8: 807–835
[7]Tai HL. Technology evaluation: Valspodar, Novartis AG. Curr Opin Mol Ther 2000; 2: 459–467
[8]Boesch D, Muller K, Pourtier-Manzanedo A et al. Restoration of daunomycin retention in multidrug-resistant P388 cells by submicro- molar concentrations of SDZ PSC 833, a nonimmunosuppressive cyclosporin derivative. Exp Cell Res 1991; 196: 26–32
[9]Lemaire M, Bruelisauer A, Guntz P et al. Dose-dependent brain penetration of SDZ PSC 833, a novel multidrug resistance-reversing cyclosporin, in rats. Cancer Chemother Pharmacol 1996; 38: 481–486
[10]Kusunoki N, Takara K, Tanigawara Y et al. Inhibitory effects of a cyclosporin derivative, SDZ PSC 833, on transport of doxorubicin and vinblastine via human P-glycoprotein. Jpn J Cancer Res 1998; 89: 1220–1228
[11]Watanabe T, Tsuge H, Oh-Hara T et al. Comparative study on reversal efficacy of SDZ PSC 833, cyclosporin A and verapamil on multidrug resistance in vitro and in vivo. Acta Oncol 1995; 34: 235–241
[12]Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem 1993; 62: 385–427
[13]Aicher L, Meier G, Norcross AJ et al. Decrease in kidney calbindin-D 28kDa as a possible mechanism mediating cyclosporine A- and
FK-506-induced calciuria and tubular mineralization. Biochem Pharmacol 1997; 53: 723–731
[14]Watanabe T, Nakayama Y, Naito M et al. Cremophor EL reversed multidrug resistance in vitro but not in tumor-bearing mouse models. Anticancer Drugs 1996; 7: 825–832

[15]Brocks DR. Stereoselective pharmacokinetics of desbutylhalofantrine, a metabolite of halofantrine, in the rat after administration of the racemic metabolite or parent drug. Biopharm Drug Dispos 2000; 21: 365–371
[16]Schuhmacher J, Buhner K, Witt-Laido A. Determination of the free fraction and relative free fraction of drugs strongly bound to plasma proteins. J Pharm Sci 2000; 89: 1008–1021
[17]Binkhathlan Z, Hamdy DA, Brocks DR et al. Pharmacokinetics of PSC 833 (valspodar) in its Cremophor EL formulation in rat. Xenobiotica 2010; 40: 55–61
[18]Binkhathlan Z, Somayaji V, Brocks DR et al. Development of a liquid chromatography-mass spectrometry (LC/MS) assay method for the quantification of PSC 833 (Valspodar) in rat plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2008; 869: 31–37
[19]Binkhathlan Z, Hamdy DA, Brocks DR et al. Development of a polymeric micellar formulation for valspodar and assessment of its pharmacokinetics in rat. Eur J Pharm Biopharm 2010; 75: 90–95
[20]Fischer V, Rodriguez-Gascon A, Heitz F et al. The multidrug resistance modulator valspodar (PSC 833) is metabolized by human cytochrome P450 3A. Implications for drug-drug interactions and pharmacological activity of the main metabolite. Drug Metab Dispos 1998; 26: 802–811
[21]Urien S, Zini R, Lemaire M et al. Assessment of cyclosporine A interactions with human plasma lipoproteins in vitro and in vivo in the rat. J Pharmacol Exp Ther 1990; 253: 305–309
[22]Jin M, Shimada T, Yokogawa K et al. Cremophor EL releases cyclosporin A adsorbed on blood cells and blood vessels, and increases apparent plasma concentration of cyclosporin A. Int J Pharm 2005; 293: 137–144
[23]van Zuylen L, Karlsson MO, Verweij J et al. Pharmacokinetic modeling of paclitaxel encapsulation in Cremophor EL micelles. Cancer Chemother Pharmacol 2001; 47: 309–318
[24]Gelderblom H, Verweij J, van Zomeren DM et al. Influence of Cremophor El on the bioavailability of intraperitoneal paclitaxel. Clin Cancer Res 2002; 8: 1237–1241
[25]Sparreboom A, van Tellingen O, Nooijen WJ et al. Nonlinear pharma- cokinetics of paclitaxel in mice results from the pharmaceutical vehicle Cremophor EL. Cancer Res 1996; 56: 2112–2115
[26]Sparreboom A, van Zuylen L, Brouwer E et al. Cremophor EL-mediated alteration of paclitaxel distribution in human blood: clinical pharma- cokinetic implications. Cancer Res 1999; 59: 1454–1457
[27]Highley MS, De Bruijn EA. Erythrocytes and the transport of drugs and endogenous compounds. Pharm Res 1996; 13: 186–195
[28]Simon N, Dailly E, Combes O et al. Role of lipoproteins in the plasma binding of SDZ PSC 833, a novel multidrug resistance-reversing cyclosporin. Br J Clin Pharmacol 1998; 45: 173–175
[29]Bagnarello AG, Lewis LA, McHenry MC et al. Unusual serum lipoprotein abnormality induced by the vehicle of miconazole. N Engl J Med 1977; 296: 497–499
[30]Kongshaug M, Cheng LS, Moan J et al. Interaction of cremophor EL with human plasma. Int J Biochem 1991; 23: 473–478
[31]Woodburn K, Kessel D. The alteration of plasma lipoproteins by cremo- phor EL. J Photochem Photobiol B 1994; 22: 197–201
[32]Kessel D, Woodburn K, Decker D et al. Fractionation of Cremophor EL delineates components responsible for plasma lipoprotein alterations and multidrug resistance reversal. Oncol Res 1995; 7: 207–212
[33]Liu B, Gordon WP, Richmond W et al. Use of solubilizers in preclinical formulations: Effect of Cremophor EL on the pharmacokinetic properties on early discovery compounds. Eur J Pharm Sci 2016; 87: 52–57