Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

Journal of Pharmaceutical and Biomedical Analysis 139 (2017) 247–251

Contents lists available at ScienceDirect

Journal of Pharmaceutical and Biomedical Analysis


journal homepage: www.elsevier.com/locate/jpba

Short Communication

HPLC–MS/MS method for quantification of paclitaxel from keratin


containing samples
Emily A. Turner a , Alexandra C. Stenson b , Saami K. Yazdani a,∗
a
Department of Mechanical Engineering, University of South Alabama, Mobile, AL 36688, USA
b
Department of Chemistry, University of South Alabama, Mobile, AL 36688, USA

a r t i c l e i n f o a b s t r a c t

Article history: Local drug delivery of paclitaxel is becoming ever more prevalent. As complex drug/excipient combina-
Received 15 December 2016 tions are being developed and tested, new high performance liquid chromatography-mass spectrometry
Received in revised form 28 February 2017 (HPLC–MS) techniques capable of quantifying paclitaxel from such formulations are needed. Here a
Accepted 9 March 2017
method for quantifying paclitaxel from aqueous, protein and oil containing samples was developed and
Available online 10 March 2017
validated. Keratin, derived from human hair, is the protein component/paclitaxel excipient in the devel-
opment and validation of said method. The novelty of this method is described by its ability to overcome
Keywords:
water solubility issues and address clean-up of residual solvents in clinical grade paclitaxel injection
Liquid chromatography
Mass spectrometry
composition. The method evaluates tert-butyl methyl ether and ethanol as extraction solvents with an
Keratin extraction efficiency of 31.9 ± 2.3% and 86.4 ± 4.5% respectively. Upon evaporation and rehydration, sam-
Keratose ples were evaluated by HPLC–MS and a method was developed for paclitaxel quantification. The method
Protein extraction developed had an inter-day precision of 9.1% relative standard deviation and an intra-day precision of
Paclitaxel 4.3% relative standard deviation normalized to a docetaxel internal standard. The described method is
applicable to any aqueous paclitaxel sample containing protein and/or oils.
© 2017 Elsevier B.V. All rights reserved.

1. Introduction With the recent approval of DCB in the US market, research


has moved to optimizing paclitaxel/excipient formulations for arte-
Paclitaxel is an anti-proliferative drug extracted from pacific rial paclitaxel delivery [10,11]. Paclitaxel excipients are capable of
yew bark that inhibits cell division by binding to growing micro- modulating coating durability, paclitaxel solubility/bioavailability,
tubules [1,2]. Used as a cancer therapeutic since 1992, paclitaxel pharmacokinetics, and vascular healing post-treatment [12–14].
is active against ovarian cancer, breast cancer, Kaposi’s sarcoma Current excipients include polysorbate and sorbitol, urea, and
and lung cancer [1,3]. More recently, paclitaxel has been used as an iopromide [15–18]. Although these excipients have improved the
anti-restenotic in cardiovascular interventions such as drug eluting success of DCBs, long-term drug residency and vascular healing
stents and drug coated balloons (DCB) to inhibit smooth muscle cell remain subpar. This opens the door for discovery of improved
proliferation − a primary contributor to restenosis [4–7]. excipients capable of elongating drug residency and improving vas-
Paclitaxel is a lipophilic drug that is nearly insoluble (0.172 mg/L cular healing in paclitaxel drug coated balloons.
at pH 7) in water, making it difficult to formulate for intra- As keratins are becoming increasingly studied in drug delivery
venous delivery [8]. Lipophilic vehicles such as polyethoxylated applications, a method is needed to quantify drug release from ker-
castor oil (Cremophor EL) are used to aid solubility. Unfortu- atins. Equivalently, methods capable of quantifying paclitaxel from
nately, Cremophor EL is toxic; it causes vasodilation, labored excipients are needed, as excipients of paclitaxel are being eval-
breathing, lethargy, and hypotension [9]. The optimized pacli- uated. Keratin is a protein extracted from hair and a widely used
taxel intravenous formulation consists of 6 mg/mL paclitaxel, biomaterial that represents potential as a novel paclitaxel excipient
polyethoxylated castor oil (Cremophor EL) and anhydrous alco- [19–21]. Keratin is highly biocompatible, has intrinsic scaffold-
hol [8]. Although the formulation of paclitaxel has been optimized, ing capability, supports release of several drugs and factors, and
solubility, precipitation, and toxicity concerns remain, highlighting promotes anti-inflammation in vivo [19,22–26]. Both keratose and
the difficulty of working with paclitaxel solutions. kerateine are accumulations of protein/protein fragments and iso-
forms of various sizes [19,22]. The hydrophilic nature and range of
molecular weights in keratin preparations complicate mass spec-
∗ Corresponding author. tral analysis.
E-mail address: syazdani@southalabama.edu (S.K. Yazdani).

http://dx.doi.org/10.1016/j.jpba.2017.03.011
0731-7085/© 2017 Elsevier B.V. All rights reserved.
248 E.A. Turner et al. / Journal of Pharmaceutical and Biomedical Analysis 139 (2017) 247–251

HPLC–MS methods describing the quantification of paclitaxel samples were stored at 4 ◦ C for no longer than one month. Qual-
from biological samples are numerous and effective in dealing ity control samples were prepared at a concentration of 0.2 ng/␮L
with many complex matrices, however a method to quantify pacli- paclitaxel. Prior to mass spectrometric analysis, all samples, includ-
taxel from oil-containing samples while maintaining mass spectral ing calibration standards and quality control samples, received an
selectivity has yet to be described [27–29]. This paper describes a addition of 0.5 ␮g docetaxel to 95 ␮L of sample to serve as the
method for paclitaxel quantification from keratose and polyethoxy- internal standard.
lated castor oil containing samples. This method can be applied to
the quantification of paclitaxel from any aqueous solution contain- 2.5. Extraction procedure
ing water-soluble protein and/or oils.
To quantify paclitaxel concentration in each sample, keratose
2. Materials and methods needed to be removed. First, samples were thawed on ice, then
200 ␮L were pipetted into a clean, siliconized, low-retention micro-
2.1. Chemicals and materials centrifuge tube. Keratose was precipitated from each sample by
the addition of ethanol. One mL of 200-proof ethanol was added to
Paclitaxel Injection, USP was purchased from Sagent Pharma- each tube. Tubes were inverted several times, sonicated (Branson
ceuticals (Schaumburg, IL). The internal standard, Docetaxel (purity Ultrasonics, Danbury, CT) for one minute and then centrifuged for
>99%), was obtained from Tocris Bioscience (Bristol, United King- 3 min at 1200 rpm. Keratose collected at the bottom of the tube. The
dom). Dimethyl sulfoxide (purity 99.7%) and high performance supernatant containing the paclitaxel was transferred to a clean
liquid chromatography (HPLC) grade methanol (purity 99.96%), tube and dried in the Vacuum Concentrator at 30 ◦ C, 1700 rpm.
water, acetonitrile (purity 99.9%) and formic acid (purity 98%) were Sample evaporation took 3–3.5 h. Dried samples were redissolved
purchased from Sigma-Aldrich (St. Louis, MO). Absolute ethanol in mobile phase. Samples were transferred to autosampler vials
200-proof (purity 99.5%) was purchased from Fisher Scientific and 0.5 ␮g of docetaxel were added as an internal standard to each
(Hampton, NH). Tert-butyl methyl ether was obtained from Acros sample.
Organics (New Jersey, US).
2.6. Selection of extraction solvent
2.2. Instrumentation
Ethanol and tert-butyl methyl ether were evaluated as extrac-
Samples were evaporated with a CentriVap Vacuum Concen-
tion solvents. Ethanol extraction was evaluated as a method
trator (Labconco, Fort Scott, KS). Separation was performed on a
for protein precipitation, and tert-butyl methyl ether extraction
Symmetry C18, 100 Å, 3.5 ␮m, 100 × 2.1 mm column (Waters Cor-
was evaluated as a method of liquid-liquid extraction. Ethanol
poration, Milford, MA) using an Accela HPLC Autosampler, Accela
extraction was performed as described above. For extraction with
600 LC Pump, and LTQ Velos Orbitrap Mass Spectrometer with Elec-
tert-butyl methyl ether, 1 mL of tert-butyl methyl ether was added
trospray Ionization (Thermo Scientific, Waltham, MA). The mass
to each sample. Tubes were inverted several times, sonicated for
spectrometer (MS) was run in positive ion mode.
1 min, and then allowed to rest for 2 min. The tert-butyl methyl
ether, organic phase was decanted into a new tube, vacuum dried,
2.3. Sample prep
and re-suspended in mobile phase for HPLC–MS.
Paclitaxel Injection, USP [6 mg/mL] was combined with phos-
phate buffered saline (PBS) (GE Healthcare Hyclone, Logan, UT) at a 2.7. Chromatographic and mass spectrometric conditions
final paclitaxel concentration of 1.43 mg/mL. Lyophilized keratose
(KeraNetics, Winston-Salem, NC) was combined with the paclitaxel Docetaxel was chosen as the internal standard based on its struc-
solution (1.43 mg/mL) as 12, 15, and 20% weight-to-volume mix- tural similarity to paclitaxel (Fig. 1). The molecular ion (m/z = 853.9
tures. Using a syringe, keratose-paclitaxel mixtures were injected for paclitaxel and m/z = 807.9 for the internal standard) was used
into 2 mL Eppendorf tubes. Tubes were centrifuged at 1200 rpm for to identify each analyte. The mobile phase consisted of acetoni-
2 min to remove bubbles and then incubated overnight at 37 ◦ C trile as mobile phase A and 0.1% formic acid in water as mobile
to form hydrogels. Each hydrogel (12, 15, and 20%) contained phase B. The following gradient was applied: 45% A (0–12 min),
500 ␮g of paclitaxel. Upon formation of the hydrogels, 500 ␮L of from 45 to 100% A (12–15 min), 100% A (15–19 min), from 100
1 x PBS were added to each tube. At time points ranging from 1 h to to 45% A (19–24 min), and 45% A (24–38 min). The flow rate for
45 days, PBS was removed and replaced with fresh PBS. PBS sam- separation was 0.2 mL/min. From minute 13–33 of the separation
ples were kept at −20 ◦ C in a clean, siliconized, low-retention tube gradient, flow was diverted to waste to prevent polyethoxylated
until ready for quantification. Prior to quantification, samples were castor oil from entering the electrospray ionization source of the
thawed on ice. Samples were diluted in starting mobile phase con- mass spectrometer. Flow was diverted back to the mass spectrom-
ditions: acetonitrile-water + formic acid 0.1% (55:45, v/v). Samples eter at 33 min, and the column was re-equilibrated with mobile
were transferred to autosampler vials (SUN-SRi, Rockwood, TN) phase for five minutes before the next injection. Sample injection
and 0.5 ␮g of docetaxel were added as an internal standard to each volume was 5 ␮L and injection solvent was methanol-water (50:50,
sample. If paclitaxel quantification was out of the linear calibration v/v) because paclitaxel and docetaxel are soluble in such a solution.
range, samples were re-diluted and re-run until they fell within the Mass spectrometer detection settings are listed in Table 1. All sam-
linear range. ples were analyzed in duplicate. Data processing was performed on
Thermo Xcalibur data system (Thermo Scientific, Waltham, MA).
2.4. Calibration and quality control samples Parameters for peak quantification were standardized for all anal-
yses.
Docetaxel was dissolved in dimethyl sulfoxide at a concen-
tration of 1 ␮g/␮L. Paclitaxel, USP (6 mg/mL) and docetaxel were 2.8. Method validation
further diluted in starting mobile phase. Six paclitaxel calibra-
tion standards were prepared at concentrations ranging from The method was evaluated for specificity, signal carryover,
0.01 ng/␮L to1.25 ng/␮L. Calibration standards and quality control intra- and inter-day precision, and calibration curve. Specificity
E.A. Turner et al. / Journal of Pharmaceutical and Biomedical Analysis 139 (2017) 247–251 249

Fig. 1. Chemical structure of (a) paclitaxel and (b) docetaxel. Paclitaxel and docetaxel have similar structures but differ in molecular weight because of the difference between
a phenyl and a tert-butyl ether attachment on the carbonyl carbon of the amide. In addition, paclitaxel also has an acteal in place of a hydroxyl sidechain in the vinyl position
on the main backbone. [Images modified from Open Chemistry Database, PubChem, NIH. CID = 36314; 148124].

Table 1 3. Results and discussion


General MS settings.

Ion Optics (V) Ethanol and TBME were compared as extraction solvents for
Multiple 00 Offset −0.04 samples containing varying concentrations of paclitaxel and ker-
Lens 0 −1.34
atose. Average extraction efficiency of samples containing keratose
Multipole 0 Offset −8.19
Lens 1 −9.53
and paclitaxel was 31.9 ± 2.3% (n = 3) extracted with TBME and
Front Lens −7.26 86.4 ± 4.5% (n = 3) for samples extracted with ethanol. This led
Back Lens −9.52 to the selection of ethanol as the extraction solvent. Extraction
Heated ESI Source efficiency of samples from which keratose was precipitated was
|Spray Voltage (kV)| 0.01
93.0 ± 3.4% (n = 32). The extraction efficiency is comparable to
Spray Current (␮A) 0.02
Source Heater Temp (◦ C) 39.23 other HPLC–MS methods for paclitaxel quantification from serum
Capillary Temp (◦ C) 300.07 which range from 73% to 98% [30–33]. Keratose concentration was
Ion Detection Settings evaluated for effect on paclitaxel extraction efficiency. At low ker-
Dynode (kV) −0.01
atose concentration (1 ␮g/␮L) paclitaxel extraction efficiency was
Multiplier 1 (V) 0.00
Multiplier 2 (V) 0.92
94.3 ± 3.4% at 4.75 ng/␮L paclitaxel and 94.1 ± 2.5% at 14.25 ng/␮L
Vacuum paclitaxel. For high keratose concentration (5 ␮g/␮L) extraction
Ion Gauge (Torr) 1.48 × 10−5 efficiency was 91.1 ± 3.9% at 4.75 ng/␮L paclitaxel and 92.4 ± 3.1%
Convection Gauge (Torr) 1.49 at 14.25 ng/␮L paclitaxel. Keratose did not significantly affect the
Main RF
paclitaxel measurement (p = 0.3987).
Main RF Detected (V) −0.00
RF Detector Temp (◦ C) 54.40 Based on previous methods [28,34–36], HPLC–MS/MS was
RF Generator Temp (◦ C) 35.66 initially performed with solutions of docetaxel [5 ng/␮L] and
paclitaxel [0.15 ng/␮L] with a mobile phase of acetonitrile-
water + formic acid 0.1% (45:55, v/v) with isocratic flow at
200 ␮L/min through a Waters Symmetry C-18 column. The gra-
dient and flow rate were adjusted to optimize peak resolution
was evaluated by comparing chromatograms of blank, ethanol- and decrease run time. Docetaxel and paclitaxel produced well-
extracted samples with paclitaxel spiked samples. Carryover was established peaks without fronting or tailing, retention times
assessed by six replicate injections of blank, ethanol-extracted sam- were 7.86 min (min) and 9.10 min respectively (Fig. 2c, d). Other
ples following an injection of a paclitaxel spiked sample. Intra-day elements that make up the paclitaxel IV solution, including
and inter-day precision were evaluated by six replicate injections polyethoxylated castor oil, eluted after the paclitaxel peak (Fig. 2a).
and evaluated as percent relative standard deviation (%RSD). The The polyethoxylated castor oil carries over into following blank,
calibration curve was formed by graphing the ratio of the peak methanol injections. To prevent this carryover, acetonitrile was
area of paclitaxel to peak area of internal standard (docetaxel) ramped up to 100% following paclitaxel and docetaxel elution and,
versus the concentration of paclitaxel. The linear region of detection concomitantly, flow was diverted to waste. Mobile phase was then
was determined by preparing a range of paclitaxel concentrations returned to starting conditions, and flow was diverted back to
between 0.001 and 150 ng/␮L. the MS. The column was re-equilibrated before the next injec-
tion. Following this method, blank/methanol injections after spiked
paclitaxel injections exhibited no carryover signal.
2.9. Statistical analysis Specificity was determined by comparing chromatograms of
blank, ethanol-extracted samples with paclitaxel spiked samples.
The data were presented as means and standard deviation. Dif- No overlapping peaks were observed.
ference of significance was set at 0.05.
250 E.A. Turner et al. / Journal of Pharmaceutical and Biomedical Analysis 139 (2017) 247–251

Fig. 2. Chromatogram of paclitaxel elution. (a and b) Elution of paclitaxel spiked solution with mobile phase acetonitrile-water + formic acid 0.1% (50:50, v/v) with a flow rate
of 400 ␮L/min. a) Total ion chromatogram, b) Selected ion chromatogram for paclitaxel (m/z = 854). (c and d) Analyte peaks with mobile phase acetonitrile-water + formic
acid 0.1% (45:55, v/v) at 200 ␮L/min. c) Selected ion chromatogram for docetaxel (m/z = 808), d) Selected ion chromatogram for paclitaxel (m/z = 854).

No carryover was detected when a blank/methanol sample was Paclitaxel release from excipients as evaluated here is relevant
injected following a docetaxel spiked sample. Following a spiked to paclitaxel delivery, specifically drug coated balloon develop-
paclitaxel injection, no paclitaxel carried over to the blank, but ment. The method has broader applications yet in the quantification
some of the late eluting additive of paclitaxel were carried over. of clinical grade paclitaxel IV injection solution through any means
Quality control (QC) samples were interspersed every five sam- of delivery. Although paclitaxel is an accepted therapeutic in the
ples at an intermediate concentration of 0.02 ng/␮L. Precision of treatment of restenosis, paclitaxel is more commonly used as a
QC samples was 12%RSD for paclitaxel and 5.4%RSD for paclitaxel cancer therapeutic. Cancer therapeutics are historically delivered
normalized to the internal standard, docetaxel. systemically, however systemic delivery leads to off-target effects
The lowest concentration of paclitaxel that could reliably be which are of significant concern, considering the toxicity of pacli-
identified was 0.01 ng/␮L. Standards extracted from keratose- taxel and Cremophor EL [9]. This has led to the exploration of
paclitaxel samples gave a linear calibration curve from 0.01 targeted drug delivery of cancer therapeutics to minimize neg-
to1.25 ng/␮L: the R2 value was 0.9992 ± 0.0004 (n = 12). ative off-target effects. As cancer treatment heads towards local
Inter-day precision for paclitaxel peak area was 13%RSD. Inter- drug delivery, methods for quantification of paclitaxel from the
day paclitaxel peak area normalized to internal standard peak tumor microenvironment will be critical in the evaluation of these
area was 9.1%RSD. Intra-day precision for paclitaxel peak area was treatments. Our method matches or improves reported methods on
7.9%RSD and intra-day precision for normalized paclitaxel peak the quantification of paclitaxel from water-soluble matrices while
area was 4.3%RSD. These%RSD values are consistent with reported achieving clean-up of residual solvents such as castor oil. These
values (between 1.04 and 12.3%RSD) in determination of paclitaxel methods are invaluable in the pursuit of localized paclitaxel deliv-
concentration from plasma samples [30–33]. ery and quantification.
E.A. Turner et al. / Journal of Pharmaceutical and Biomedical Analysis 139 (2017) 247–251 251

4. Conclusions [14] W. Kempin, S. Kaule, T. Reske, N. Grabow, S. Petersen, S. Nagel, K.P. Schmitz,
W. Weitschies, A. Seidlitz, In vitro evaluation of paclitaxel coatings for delivery
via drug-coated balloons, Eur. J. Pharm. Biopharm. 96 (2015) 322–328.
A protocol for removing solutes which are insoluble in organic [15] W.A. Gray, J.F. Granada, Drug-coated balloons for the prevention of vascular
solvents and quantifying paclitaxel from the complex, paclitaxel restenosis, Circulation 121 (2010) 2672–2680.
IV injection solution (Sagent Pharmaceuticals) using HPLC–MS [16] B. Kelsch, B. Scheller, M. Biedermann, Y.P. Clever, S. Schaffner, D. Mahnkopf, U.
Speck, B. Cremers, Dose response to Paclitaxel-coated balloon catheters in the
was developed. The method was validated based on linear range porcine coronary overstretch and stent implantation model, Invest. Radiol. 46
(0.01 to1.25 ng/␮L) and intra- and inter-day precision (4.3 and (2011) 255–263.
7.9% respectively). As complex paclitaxel/excipient combinations [17] D. Scheinert, S. Duda, T. Zeller, H. Krankenberg, J. Ricke, M. Bosiers, G. Tepe, S.
Naisbitt, K. Rosenfield, The LEVANT I (Lutonix paclitaxel-coated balloon for
become more prevalent in the study of local drug delivery, this
the prevention of femoropopliteal restenosis) trial for femoropopliteal
method will allow the evaluation of drug elution in vitro. The revascularization: first-in-human randomized trial of low-dose drug-coated
authors have no conflicts of interest. balloon versus uncoated balloon angioplasty, JACC Cardiovasc Interv. 7 (2014)
10–19.
[18] A. Posa, N. Nyolczas, R. Hemetsberger, N. Pavo, O. Petnehazy, Z. Petrasi, G.
Funding Sangiorgi, M. Gyongyosi, Optimization of drug-eluting balloon use for safety
and efficacy: evaluation of the 2nd generation paclitaxel-eluting DIOR-balloon
in porcine coronary arteries, Catheter. Cardiovasc. Interv. 76 (2010) 395–403.
This study was supported by American Heart Association
[19] G.D. Mogosanu, A.M. Grumezescu, M.C. Chifiriuc, Keratin-based biomaterials
[#15SDG25880000, #16PRE27350003], National Institute of Health for biomedical applications, Curr. Drug Targets 15 (2014) 518–530.
[#1R15HL127596] and internal funding from the University of [20] T.R. Ham, R.T. Lee, S. Han, S. Haque, Y. Vodovotz, J. Gu, L.R. Burnett, S.
South Alabama. Tomblyn, J.M. Saul, Tunable keratin hydrogels for controlled erosion and
growth factor delivery, Biomacromolecules 17 (2016) 225–236.
[21] M. Curcio, B. Blanco-Fernandez, L. Diaz-Gomez, A. Concheiro, C.
Acknowledgements Alvarez-Lorenzo, Hydrophobically modified keratin vesicles for
GSH-responsive intracellular drug release, Bioconjug. Chem. 26 (2015)
1900–1907.
The authors would like to thank Shelby Boyd, BS for her help in [22] P. Hill, H. Brantley, M. Van Dyke, Some properties of keratin biomaterials:
optimizing mass spectrometer settings. kerateines, Biomaterials 31 (2010) 585–593.
[23] R.C. de Guzman, M.R. Merrill, J.R. Richter, R.I. Hamzi, O.K. Greengauz-Roberts,
M.E. Van Dyke, Mechanical and biological properties of keratose biomaterials,
References Biomaterials 32 (2011) 8205–8217.
[24] L.R. Burnett, M.B. Rahmany, J.R. Richter, T.A. Aboushwareb, D. Eberli, C.L.
[1] M.C. Wani, H.L. Taylor, M.E. Wall, P. Coggon, A.T. McPhail, Plant antitumor Ward, G. Orlando, R.R. Hantgan, M.E. Van Dyke, Hemostatic properties and the
agents VI. Isolation and structure of taxol, a novel antileukemic and antitumor role of cell receptor recognition in human hair keratin protein hydrogels,
agent from Taxus brevifolia, J. Am. Chem. Soc. 93 (1971) 2325–2327. Biomaterials 34 (2013) 2632–2640.
[2] E.K. Rowinsky, R.C. Donehower, Paclitaxel (Taxol), N. Engl. J. Med. 15 (1995) [25] J.M. Saul, M.D. Ellenburg, R.C. de Guzman, M. Van Dyke, Keratin hydrogels
1004–1014. support the sustained release of bioactive ciprofloxacin, J. Biomed. Mater. Res.
[3] E.K. Rowinsky, Clinical pharmacology of taxol, J. Natl. Cancer Inst. Monogr. 15 A 98 (2011) 544–553.
(1993) 25–37. [26] B.V. Fearing, M.E. Van Dyke, In vitro response of macrophage polarization to a
[4] M. Oberhoff, C. Herdeg, A. Baumbach, K.R. Karsch, Stent-based antirestenotic keratin biomaterial, Acta Biomater. 10 (2014) 3136–3144.
coatings (sirolimus/paclitaxel), Catheter. Cardiovasc. Interv. 55 (2002) [27] J.J. Hendrikx, H. Rosing, A.H. Schinkel, J.H. Schellens, J.H. Beijnen,
404–408. Quantification of taxanes in biological matrices: a review of bioanalytical
[5] J.P. Loh, R. Waksman, Paclitaxel drug-coated balloons: a review of current assays and recommendation for development of new assays, Bioanalysis 6
status and emerging applications in native coronary artery de novo lesions, (2014) 993–1010.
JACC Cardiovasc. Interv. 5 (2012) 1001–1012. [28] J. Jones, L. Denbigh, LC–MS/MS Method for the Determination of Paclitaxel in
[6] D.I. Axel, W. Kunert, C. Goggelmann, M. Oberhoff, C. Herdeg, A. Kuttner, D.H. Human Serum, 2013.
Wild, B.R. Brehm, R. Riessen, F. Koveker, K.R. Karsch, Paclitaxel inhibits [29] G.K. Poon, J. Wade, J. Bloomer, S.E. Clarke, J. Maltas, Rapid screening of taxol
arterial smooth muscle cell proliferation and migration in vitro and In vivo metabolites in human microsomes by liquid chromatography/electrospray
using local drug delivery, Circulation 96 (1997) 636–645. ionization–mass spectrometry, Rapid Commun. Mass Spectrom. 10 (1996)
[7] I. Haehnel, E. Alt, A. Resch, A. Märkl, A. Stemberger, I. Schömig, Local growth 1165–1168.
inhibitory effect of paclitaxel released by a biodegradable stent coating on [30] E.R. Gardner, W. Dahut, W.D. Figg, Quantitative determination of total and
vascular smooth muscle cells, J. Am. Coll. Cardiol. 31 (1998) 278A (abstract no. unbound paclitaxel in human plasma following Abraxane treatment, J.
1114-102). Chromatogr. B Anal. Technol. Biomed. Life Sci. 862 (2008) 213–218.
[8] M. Suffness, TAXOL Science and Applications, CRC Press, Boca Raton, FL, 1995. [31] H. Lian, J. Sun, T. Zhang, A rapid and sensitive determination of paclitaxel in
[9] Bristol-Myers Squibb Company, Material Safety and Data Sheet: TAXOL rat plasma by UPLC–MS/MS method: application to a pharmacokinetic study,
Injection, 2017, Retrieved April 8, 2016 from: https://secure.mypss.com/ Asian J. Pharm. Sci. 8 (2013) 199–205.
catMaintDataServlet?existing.FileId=15434. [32] R.K. Tekade, A. D’Emanuele, A. Elhissi, A. Agrawal, A. Jain, B.T. Arafat, N.K. Jain,
[10] U.S. Food and Drug Administration, Center for Devices and Radiological Extraction and RP-HPLC determination of taxol in rat plasma, cell culture and
Health. Lutonix 035 Drug Coated Balloon PTA Catheter P130024 approval quality control samples, J. Biomed. Res. 27 (2013) 394–405.
letter, October 9, 2014. Retrieved February 1, 2017, from: http://www. [33] M. Rezazadeh, J. Emami, A. Mostafavi, M. Rostami, F. Hassanzadeh, H. Sadeghi,
accessdata.fda.gov/cdrh docs/pdf13/P130024a.pdf. M. Minaiyan, A. Lavasanifar, A rapid and sensitive HPLC method for
[11] U.S. Food and Drug Administration, Center for Devices and Radiological quantitation of paclitaxel in biological samples using liquid-liquid extraction
Health. IN.PACTTM AdmiralTM Paclitaxel-coated Percutaneous Transluminal and UV detection: paclitaxel loaded targeted polymeric micelles in tumor
Angioplasty (PTA) Balloon Catheter P140010 approval letter, December 30, bearing mice, J. Pharm. Pharm. Sci. 18 (2015) 647–660.
2014. Retrieved February 1, 2017, from: http://www.accessdata.fda.gov/cdrh [34] M.C. Berg, H. Kolodziej, B. Cremers, G. Gershony, U. Speck, Drug-coated
docs/pdf14/P140010a.pdf. angioplasty balloon catheters: coating compositions and methods, Adv. Eng.
[12] N. Lockwood, Drug delivery to the vessel wall: coated balloons and the role of Mater. 14 (2012) B45–B50.
the excipient, BioInterface (2015), Retrieved March 7, 2016 from: http://c. [35] U. Speck, B. Cremers, B. Kelsch, M. Biedermann, Y.P. Clever, S. Schaffner, D.
ymcdn.com/sites/www.surfaces.org/resource/collection/4423FA75-D640- Mahnkopf, U. Hanisch, M. Bohm, B. Scheller, Do pharmacokinetics explain
4955-A412-240A38EF1FAA/2015 Nathan Lockwood.pdf. persistent restenosis inhibition by a single dose of paclitaxel? Circ Cardiovasc.
[13] A. Seidlitz, N. Kotzan, S. Nagel, T. Reske, N. Grabow, C. Harder, S. Petersen, K. Interv. 5 (2012) 392–400.
Sternberg, W. Weitschies, In vitro determination of drug transfer from [36] J.B. Wolinsky, Y.L. Colson, M.W. Grinstaff, Local drug delivery strategies for
drug-coated balloons, PLoS One 8 (2013) e83992. cancer treatment: gels, nanoparticles, polymeric films, rods, and wafers, J.
Control. Release 159 (2012) 14–26.

You might also like