ArticlePDF Available

Chemical Modifications of Therapeutic Proteins Induced by Residual Ethylene Oxide

Authors:

Abstract and Figures

Ethylene oxide (EtO) is widely used in sterilization of drug product primary containers and medical devices. The impact of residual EtO on protein therapeutics is of significant interest in the biopharmaceutical industry. The potential for EtO to modify individual amino acids in proteins has been previously reported. However, specific identification of EtO adducts in proteins and the effect of residual EtO on the stability of therapeutic proteins has not been reported to date. This paper describes studies of residual EtO with two therapeutic proteins, a PEGylated form of the recombinant human granulocyte colony-stimulating factor (Peg-GCSF) and recombinant human erythropoietin (EPO) formulated with human serum albumin (HSA). Peg-GCSF was filled in an EtO sterilized delivery device and incubated at accelerated stress conditions. Glu-C peptide mapping and LC–MS analyses revealed residual EtO reacted with Peg-GCSF and resulted in EtO modifications at two methionine residues (Met-127 and Met-138). In addition, tryptic peptide mapping and LC–MS analyses revealed residual EtO in plastic vials reacted with HSA in EPO formulation at Met-328 and Cys-34. This paper details the work conducted to understand the effects of residual EtO on the chemical stability of protein therapeutics. © 2014 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci
Content may be subject to copyright.
RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology
Chemical Modifications of Therapeutic Proteins Induced by
Residual Ethylene Oxide
LOUISE CHEN, CHRISTOPHER SLOEY, ZHANG ZHONGQI, PAVEL V. BONDARENKO, HYOJIN KIM, DA REN, SEKHAR KANAPURAM
Process and Product Development, Amgen Inc., Thousand Oaks, California
Received 10 September 2014; revised 13 October 2014; accepted 17 October 2014
Published online in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.24257
ABSTRACT: Ethylene oxide (EtO) is widely used in sterilization of drug product primary containers and medical devices. The impact of
residual EtO on protein therapeutics is of significant interest in the biopharmaceutical industry. The potential for EtO to modify individual
amino acids in proteins has been previously reported. However, specific identification of EtO adducts in proteins and the effect of
residual EtO on the stability of therapeutic proteins has not been reported to date. This paper describes studies of residual EtO with two
therapeutic proteins, a PEGylated form of the recombinant human granulocyte colony-stimulating factor (Peg-GCSF) and recombinant
human erythropoietin (EPO) formulated with human serum albumin (HSA). Peg-GCSF was filled in an EtO sterilized delivery device and
incubated at accelerated stress conditions. Glu-C peptide mapping and LC–MS analyses revealed residual EtO reacted with Peg-GCSF
and resulted in EtO modifications at two methionine residues (Met-127 and Met-138). In addition, tryptic peptide mapping and LC–MS
analyses revealed residual EtO in plastic vials reacted with HSA in EPO formulation at Met-328 and Cys-34. This paper details the work
conducted to understand the effects of residual EtO on the chemical stability of protein therapeutics. C2014 Wiley Periodicals, Inc. and
the American Pharmacists Association J Pharm Sci
Keywords: drug delivery systems; injectables; protein delivery; chromatography; HPLC; mass spectrometry; proteins; peptides; formula-
tion; analytical biochemistry
INTRODUCTION
Common sterilization methods include moist heat (i.e., steam),
dry heat, ionizing radiation (gamma or e-beam), hydrogen per-
oxide, and ethylene oxide (EtO). Each method has its advan-
tages and limitations. Ionizing radiation, hydrogen peroxide,
and EtO often leave residuals that are damaging to proteins
and DNA.1–4 EtO sterilization is particularly useful for steriliz-
ing delicate medical devices and heat and/or moisture sensitive
equipments or materials.5,6 However, the difficulties caused by
the potential hazards of EtO to patients, staff, and the envi-
ronment as well as risk associated with handling a flammable
gas make EtO sterilization a challenging task.5Because dif-
ferent materials adsorb and desorb EtO differently,7the allow-
able levels of residual EtO remaining in sterilized apparatus
and materials need to be specified for individual products per
regulations. Therefore, the sterilization process, including EtO
concentration, sterilization time, temperature, and humidity
as well as the aeration process conditions and time must be
optimized for effectively eliminating residual EtO.8
Although EtO has been utilized for decades, protein degra-
dation by EtO has not been widely reported in the literature. An
Abbreviations used: ACN, acetonitrile; CEX, cation-exchange chromatog-
raphy; CID, collision-induced dissociation; DTT, dithiothreitol; EDTA,
ethylenediaminetetraacetic acid; EPO, recombinant human erythropoietin;
EtO, ethylene oxide; GC/FID, gas chromatography equipped with flame ion-
ization detector; GC–MS, gas chromatography coupled with MS; Gdn-HCl,
guanidine-HCl; HSA, human serum albumin; IAA, iodoacetic acid; LC–MS,
liquid chromatography coupled with MS; MS/MS, fragmentation mass spectra;
Peg-GCSF, pegylated recombinant human granulocyte colony-stimulating factor;
PFS, prefilled syringe; PP1, post peak 1; PP2, post peak 2; PP3, post peak 3;
RP-HPLC, reversed-phase HPLC; RT, retention time; TFA, trifluoroacetic
acid.
Correspondence to: Louise Chen (Telephone: +805-447-6488; Fax: +805-376-
8505; E-mail: louise.chen@amgen.com)
Journal of Pharmaceutical Sciences
C
2014 Wiley Periodicals, Inc. and the American Pharmacists Association
initial model system found it forms adducts with methionine,
cysteine, and histidine amino acid residues. The model experi-
ments conducted by reaction of EtO with commercial proteins
also reported a decrease in biological activity. The reduction in
bioactivity appeared to be correlated with the electrophilic hy-
droxyethylation of an atom with one or more lone pairs of elec-
trons, particularly nitrogen and sulfur.1Gas chromatography
coupled with MS (GC–MS) and liquid chromatography coupled
with MS (LC–MS) were applied in measuring small-molecule
DNA and protein adducts released during EtO treatments,
such as N-(2-hydroxyethyl) valine, N1-(2-hydroxyethyl) histi-
dine, and S-(2-hydroxyethyl) cysteine.4Thelackofinformation
about tandem MS (MS/MS) analysis of EtO-modified peptides
in proteins made site identification in protein sequence and
quantitation of the modification analytically challenging. The
challenges were because of the poor gas-phase fragmentation
of peptides containing methionine residues modified by EtO. To
date, identification of this modification is not widely described
in the literature.
In this report, detection and identification of the protein
degradation caused by residual EtO (low levels of EtO remain-
ing in the different medical containers) using two therapeutic
proteins, a PEGylated form of recombinant human granulo-
cyte colony-stimulating factor (Peg-GCSF), and a recombinant
human erythropoietin (EPO) formulated with human serum
albumin (HSA), were studied. EtO sterilized delivery devices
were filled with Peg-GCSF from a prefilled syringe (PFS) for-
mulated at 10 mg/mL in 10 mM sodium acetate, 5% sorbitol,
pH 4.0, 0.004% polysorbate 20 and incubated at an accelerated
condition of 42 ±1C for 54 h. Samples were analyzed by sev-
eral analytical assays to evaluate the impact of residual EtO
on Peg-GCSF. Assay results showing the impact of EtO, in-
cluding cation-exchange chromatography/HPLC (CEX–HPLC)
and reversed-phase HPLC (RP-HPLC) are highlighted in this
Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES 1
2RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology
Figure 1. Identification of all minor species present in the retention time window of 40–56 min in Glu-C peptide map of Peg-GCSF. Blue trace,
filled device control; red trace, stressed filled device; green trace, PFS control; pink trace, stressed PFS.
report. Endoproteinase Glu-C digested peptide maps analyzed
by LC/MS were used for identification and characterization of
any EtO modifications.
In a separate experiment, EPO was formulated at
0.0168 mg/mL in 20 mM sodium citrate buffer pH 6.9 with
100 mM sodium chloride and 2.5 mg/mL HSA. A 1.0 mL sam-
ple was filled in plastic vials which had been custom sterilized
using an EtO process. Vials were incubated at 37 ±1Cforup
to 2 months after which they were analyzed by several assays
to evaluate the impact of residual EtO on EPO with HSA.
EXPERIMENTAL PROCEDURES
Materials and Equipment
Chemicals and Materials
Sodium acetate, sodium chloride, glycerol, trifluoroacetic
acid (TFA), tris(hydroxymethyl)aminomethane (Tris), Tris
hydrochloride (Tris–HCl), urea, hydroxylamine, dithiothre-
itol (DTT), iodoacetic acid (IAA), ethylenediaminetetraacetic
acid (EDTA), and guanidine-HCl (Gdn-HCl) were ACS grade
(Sigma–Aldrich, St. Louis, Missouri). All organic solvents were
of analytical or HPLC grade. Trypsin and Endoproteinase Glu-
C was Roche sequencing grade (Roche Diagnostics Corporation,
Indianapolis, Indinana). Vydac C-4 columns (250 ×2.1 mm2,
5-:m particle size) were purchased from Grace Co. (Columbia,
Maryland). PLRS-S columns (4.6 ×150 mm2)andVarianPo-
laris C18 Ether columns (2.0 ×250 mm2,3-:mparticlesize)
were purchased from Agilent Technologies (Santa Clara, Cal-
ifornia). Cation exchange columns (TSK gel SP-NPR, 4.6 ×
35 mm2, 2.5-:m nonporous particle) were from TosoHaas Bio-
science Company (Phenomenex, part # CHO-8039; King of
Prussia, Pennsylvania). Delivery devices were manufactured
from Insulet Company (USA).
Instruments
A Thermo Scientific (Waltham, Massachusetts) Velos Ion Trap
(LTQ) mass spectrometer was used for Glu-C peptide map-
ping analysis and CEX post peak characterization and an Ag-
ilent 1100 series HPLC was used for the RP-HPLC assay. A
Dionex (Sunnyvale, California) Ultimate 3000 HPLC was used
for the CEX–HPLC assay. Agilent headspace gas chromatogra-
phy equipped with flame ionization detector (FID) was used for
EtO analysis.
Protein Formulations
Peg-GCSF (pegfilgrastim) was formulated at 10 mg/mL in
10 mM sodium acetate, 5% sorbitol, pH 4.0, and 0.004% polysor-
bate 20. Epoetin alfa (recombinant human EPO) was formu-
lated at 0.0168 mg/mL in 20 mM sodium citrate buffer (pH 6.9)
containing 100 mM sodium chloride and 2.5 mg/mL HSA.
Methods
EtO-Degraded Peg-GCSF Sample Preparation
Peg-GCSF was loaded into an EtO sterilized delivery device
from a glass PFS. The filled devices along with the initial PFS
were subjected to storage at an accelerated condition of 42 ±
1C for 54 h. The initial PFS was known to contain no residual
EtO and served as a control at the stressed condition. Post
incubation, the Peg-GCSF in the stressed containers and PFS
control samples were each collected for testing. The PFS and
filled containers at time zero (unstressed) were also included as
controls. All samples were digested with endoproteinase Glu-C
Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24257
RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology 3
Figure 2. The fragmentation mass spectrum (MS/MS) of the 4+ion with m/z1019.2 (43.6 min) and the 3+ion with m/z1357.9 (46.2 min)
suggested a 92 Da loss in the side chain during collision-induced dissociation (CID).
Figure 3. MS/MS for the 4072 Da mass peak at 43.6 min (4+ion of m/z1019.2) shown in the square root ion intensity scale was used to assign
the peaks and identify the site of the modification in G7 native peptide (L125-E163).
followed by analysis using a RP-HPLC coupled to the Velos Ion
Trap mass spectrometer.
Glu-C Peptide Mapping
Peg-GCSF test samples were digested with endoproteinase Glu-
C in digestion buffer [0.5 M Tris, 0.5 M Tris–HCl, 0.4 M methy-
lamine hydrochloride (CH3NH2·HCl), 0.2 M DTT, and 6 M urea]
and incubated at 25C for 18 h. A Vydac C-4 (250 ×2.1 mm2,
5-:m particle) reversed phase column was used and maintained
at 40C with a flow rate of 0.2 mL/min. Mobile phase A was
0.1% TFA in water. Mobile phase B was 0.09% TFA in 90%
acetonitrile (ACN). Peptides were separated by ramping the
mobile phase B from 2% to 60% over 75 min and followed by
column cleanup and system equilibration. The total run time is
DOI 10.1002/jps.24257 Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
4RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology
Scheme 1. EtO and methionine adduct formation in the G7 peptide
(L125-E163) of Peg-GCSF and the process for gas-phase side-chain loss.
120 min. Eluted peptides were detected by UV at 214 nm, fol-
lowed by mass spectrometric analysis.
CEX–HPLC Analysis
A SP-NPR (4.6 ×35 mm2, 2.5-:m nonporous particles) CEX
column was used and maintained at 25Cwithaflowrateof
1.0 mL/min. Mobile phase A was 5 mM sodium acetate and 5%
glycerol, pH 5.6. Mobile phase B was 5 mM sodium acetate,
100 mM NaCl, and 5% glycerol, pH 5.6. The separation of the
charged variants was carried out by ramping the mobile phase
B from 0% to 33% over 60 min.
Tryptic Peptide Mapping of EPO/HSA
Each EPO sample containing 0.5 mg HSA was diluted in a
solution containing 7.5 M Gdn-HCl, 0.25 M Tris, and 2 mM
EDTA. Then, 30 :L of 0.5M DTT was added and sample was
incubated at room temperature for 30 min. Next, 7 :Lof
0.5 M IAA was added and samples were incubated for 15 min in
the dark. Then, 4 :L of 0.5 M DTT was added to stop the alky-
lation reaction. Samples were desalted using NAP-5 columns
(GE Healthcare, Piscataway, New Jersey) prior to adding of re-
combinant trypsin solution (Roche). Incubation was conducted
at 37C for 30 min. Mobile phase A was 0.1% TFA in water.
Mobile phase B was 0.085% TFA in 90% ACN. The HPLC col-
umn used was a Varian Polaris Ether, C18, 2.0 mm ID ×250
mm length, 3-:m particle size, with a 300 ˚
A pore size. The col-
umn temperature was maintained at 50C, and the flow rate
was 0.2 mL/min. Peptides were separated by ramping mobile
phase B from 0% to 50% over 195 min. Detection of peptides
was performed using a UV detector set at 214 nm, followed by
mass spectrometric analysis using the LTQ Velos instrument.
The mass spectrometric conditions were at positive mode, full
scan followed by a zoom scan and MS/MS with collision-induced
dissociation (CID) scans, with dynamic exclusion.
RESULTS
EtO Analysis by Gas Chromatography
A delivery device with EtO sterilized in a cGMP like process
was used as the residual EtO source. The residual EtO level
in the experimental devices were quantified at approximately
40 :g based on the analysis by headspace gas chromatography
equipped with FID (GC/FID).9Similarly, the residual EtO level
in the sterilized plastic vials were determined approximately
30 :g per vial.
Detection and Identification of EtO Modification in Peg-GCSF
The LC/UV traces in the stressed filled device and controls from
the Glu-C peptide mapping analysis were overlaid for compari-
son (Fig. 1). No differences were observed for the major peptides
(G1–G8) in the Glu-C peptide map. However, in the selected re-
tention time (RT) window of 40–56 min (zoom scale), two minor
species at 43.6 and 46.2 min showed a significant increase in
the filled device samples. The peak at 43.6 min [ions: m/z1358.0
(3+), m/z1019.2 (4+), and m/z1354.4 (3+)] was identified as a
coelution of methionine-oxidized peptide L125-E163 (contain-
ing methionine oxidation at both sites of Met-127 and Met-138,
mass 4060 Da)10,11 and an unknown species (mass 4072 Da).
The peak at 46.2 min [ions: m/z1357.9 (3+), m/z1019.0 (4+),
and 1128.5 (2+)] was identified as a coelution of Met-122Ox
Figure 4. CEX profiles of Peg-GCSF for stressed filled device compared with PFS and device controls. Black trace, PFS control; blue trace, filled
device control; pink trace, stressed PFS; brown trace, stressed filled device.
Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24257
RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology 5
Figure 5. Characterization of the CEX–HPLC fractions by Glu-C peptide map. (a) No modified species were found in the main peak fraction.
(b) The PP1 fraction was enriched with Met-127 EtO adducts. (c) The PP3 fraction was enriched with Met-138 EtO adducts. (d) The PP2 fraction
was a mixture of various oxidized methionines. Peak (#1) represents bothOx (OxMet-127 and OxMet-138) for the G7 peptide (L125-E163) with a
monoisotopic mass of 4057 Da. Peak (#2) represents OxMet-138 for the G7 peptide with a monoisotopic mass 4041 Da. Peak (#3) represents OxMet-
122 for the peptide T106-E124 with a monoisotopic mass of 2254 Da. Peak (#4) represents OxMet-127 for the G7 peptide with a monoisotopic
mass of 4041 Da. Peak (#5) represents OxMet-122 for the G6 peptide (L100-E124) with a monoisotopic mass of 2850 Da. Several minor species
were observed in the overlaid traces of (b) and (c) and the sources may be because of the impurities from the neighboring peaks in the collected
fractions of PP1 and PP3.
(methionine sulfoxide formed at methionine-122) for peptide
T106-E124 (mass 2255 Da) and an unknown species (mass
4072 Da).
To identify the unknown 4072 Da species, a Thermo Scien-
tific LTQ Velos ion trap mass spectrometer was operated in full
scan (MS) mode followed by a fragmentation scan (MS/MS). The
MS/MS spectra of the 4+ion with m/z1019.2 (43.6 min) and
3+ion with m/z1357.9 (46.2 min) were both found to contain
an unusual peptide fragmentation pattern. MS/MS of each ion
resulted in a single major fragment indicating a loss of 92 Da
from the 4072 Da species (Fig. 2) inferring that an EtO-modified
side chain of an amino acid residue may be susceptible to losing
the modified side chain with a 92 Da loss (CH3SCH2CH2OH)
during CID. A similar fragmentation pattern was reported for
multiply charged ions containing oxidized methionine12,13 in-
dicating that the modified side chain of methionine is suscep-
tible to fragmentation and is easily loss of 64 Da (CH3SOH).
It was noticed that the expected m/zare 1018.9 Da (4+)and
1358.2 Da (3+) (theoretical average mass 4071.6 Da), which
differs to the measured m/zby 0.3 amu approximately. This
difference is common for low-resolution spectrum of multiply
charged ions in Velos ion trap Mass Spectrometer.
The MS/MS confirmed that the 4072 Da species at 43.6 and
46.2 min correspond to peptide L125-E163 with EtO-modified
methionine side chains at Met-138 and Met-127, respectively.
Figure 3 showed the MS/MS and fragment assignments of the
4+ion at 43.6 min for Met-138. To magnify the low-abundance
fragments, the MS/MS spectra were displayed in square-root
scale. Data for Met-127 are not shown. An illustration of the
EtO and methionine reaction as well as gas-phase side-chain
loss is shown in Scheme 1.
Characterization of the Post Peaks Found by CEX–HPLC
The Peg-GCSF that had been stressed at an elevated tempera-
ture in the device displayed a different CEX profile when com-
pared with the controls. Three post peaks were found in the
stressed samples by CEX–HPLC. The impurities of post peaks
in the stress samples were quantified approximately 4.4%–5.8%
(Fig. 4). Where post peak #1 (PP1) increased relative to controls
and post peaks #2 and #3 (PP2 and PP3) were new peaks. These
two new peaks were neither found in historical stability data
nor in previous forced degradation studies for Peg-GCSF. It was
suspected that these post peaks were related to the residual
EtO in the stressed device samples.
DOI 10.1002/jps.24257 Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
6RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology
Figure 6. Tryptic peptide map of EPO showed two EtO modifications in EtO sterilized plastic vial (green) when compared with the unsterilized
plastic (red) and glass vial (blue) controls.
Figure 7. MS/MS for m/z834 at RT 131 min indicates a 44 Da mass increase of the parent ion resulting from ETO reaction with Met-328 of
HSA peptide D324-R336 followed by loss of the modified methionine side chain (92 Da) from fragment ions.
To characterize the three CEX post peaks, fractions were col-
lected from the CEX elution using an analytical CEX column,
SP-NPR (4.6 ×35 mm2, 2.5-:m nonporous particles). The frac-
tions included the main peak and the three post peaks (PP1,
PP2, and PP3). Each collected fraction was concentrated and
buffer-exchanged with product formulation buffer containing
no polysorbate. The main peak fraction contained the expected
intact molecule without any protein modifications and was used
as a study control for the entire collection and characterization
process. The post peak fractions (PP1, PP2, and PP3) were rel-
atively low in concentration, and therefore were each added
to the PFS sample (which contained no residual EtO) to make
up the final sample concentration needed for Glu-C peptide
mapping analysis. These solutions were then verified by CEX–
HPLC to confirm the identity and purity prior to performing the
characterization. The CEX–HPLC traces confirm the integrity
of the fraction collecting process performed on the starting Peg-
GCSF material from the stressed devices (data not shown).
The PFS-fraction solutions were then each digested by Glu-
C endoproteinase followed by reversed-phase LC–MS analyses.
The LC–UV traces from individual digested solutions were each
overlaid with the digested solution from PFS only to identify the
major changes in the Glu-C peptide profile. No changes were
identified in the chromatogram of the main peak fraction Glu-C
Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24257
RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology 7
Figure 8. HPLC–UV chromatogram overlay. The 14 Da difference between two peaks at 162 and 163 min is from +44 EtO modification of
Cys-34 in the absence of the +58 modification from alkylation.
digest, which confirmed that the collection and characterization
process had no impact (Fig. 5a). One major change was seen in
the PP1 fraction Glu-C digest (RT: 45.4 min) which was identi-
fied as being enriched with Met-127 EtO adducts based on its
RT (Fig. 5b), mass and MS/MS data (data not shown). Simi-
larly, the major change seen in the PP3 fraction Glu-C digest
(RT: 42.9 min) was identified as enriched Met-138 EtO adducts
(Fig. 5c). Note that several minor changes were also observed
in PP1 and PP3 fraction Glu-C digests, which may be because
of the impurities from adjacent peaks in the collected fractions.
For the PP2 fraction Glu-C digest, multiple peaks were found
with higher intensity in the overlaid UV traces. These peaks
were identified as several oxidized methionine residues (Met-
138, Met-127, and Met-122) (Fig. 5d).
The CEX fractions were also tested by an intact protein
RP-HPLC method to compare the hydrophobicity of the EtO
adducts. The RP-HPLC traces indicated that all post peak frac-
tion chromatograms are eluted at the prepeaks region on the
RP-HPLC. The fractions PP1 (Met-127 EtO adducts) and PP3
(Met-138 EtO adducts) were eluted earlier than fraction PP2
(multiple oxidation species) and the main peak (no modifica-
tion). This indicates that the EtO adducts are less hydrophobic
than the oxidation species and main peak, presumably because
of the introduction of the charged side chain.
Detection and Identification of EtO-Induced Modifications of
HSA in EPO Formulations
Erythropoietin samples containing HSA that had been incu-
bated at 37C for 2 weeks were digested with trypsin and an-
alyzed by LC–MS. Samples that had been incubated in EtO-
sterilized plastic vials were compared with control samples that
had been incubated in unsterilized plastic vials as well as in
heat-depyrogenated glass vials. Figure 6 shows the HPLC–UV
chromatogram comparison for these samples. Two peaks are
shown to be significantly different in the EtO sterilized vial,
one at a RT of 131 min and another at 163 min. From the MS
analysis, the peak at 131 min has a mass of 1666.72 Da and
was speculated to be indicative of a 44 Da increase resulting
from EtO reaction with Met-328 of HSA peptide D324-R336.
Met-328 showed susceptibility to oxidation indicating that it is
solvent exposed on the surface of HSA (data not shown). Similar
to the previously described on Peg-GCSF results, the MS/MS
results for this EtO-modified peptide indicate a predominant
fragmentation pattern resulting in a loss of 92 Da because of
the loss of the modified side chain of methionine. Analysis of
lower abundance fragmentation products of mass spectrum re-
vealed fragments that confirmed the identity of the modified
peptide as D324-R336 of HSA (Fig. 7).
Close examination of the HPLC–UV chromatogram revealed
that the increase in the new peak at 163 min in the EtO-
sterilized plastic vial sample coincided with the decrease of
the adjacent peak eluting 1 min earlier. The native peak elut-
ing at 162 min was identified by MS/MS as A21-K41 of HSA
with a +58 Da modification of Cys-34 because of the reduction
and alkylation procedure performed prior to the tryptic digest
(Fig. 8). This peptide does not contain a methionine residue.
However, a previously published report indicated the possibil-
ity of cysteine reacting with EtO.1In addition, Cys-34 of HSA
is known to be the only free cysteine residue in HSA, with the
other 34 cysteine residues in HSA forming 17 disulfide bonds.14
DOI 10.1002/jps.24257 Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
8RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology
It was assumed that EtO adduct formation on Cys-34 would
prevent its alkylation during sample preparation and would
result in a 14 Da change in the expected mass of the Cys-
containing peptide. Comparing the full scan MS as well as the
MS/MS spectra revealed that the two peptides (RT: 162 and
163 min) are closely related but with a difference in mass of
14 Da. The native peptide with cysteine alkylation has a mass
of 2491 Da, whereas the new peak in the EtO sample has a mass
of 2477 Da. The MS/MS spectrum for the new peak is identi-
fied as a +44 Da modification of Cys-34 in the absence of the
+58 Da modifications from alkylation thus accounting for the
14 Da difference between the two peptides. The MS/MS frag-
mentation pattern of the EtO-modified cysteine peptide did not
show the side-chain loss pattern displayed by the EtO-modified
methionine peptides (data not shown).
Erythropoietin is somewhat unique because of the fact that
its formulation contains a second protein. HSA is added as a
stabilizer and is present at a significantly higher concentra-
tion relative to EPO and is therefore expected to be the greater
scavenger for side-chain modifiers. Thus, the relatively low con-
centration of EPO in the formulation, EtO-modified EPO could
not be detected in the tryptic peptide map performed in this
study.
DISCUSSION AND CONCLUSION
The Glu-C and tryptic peptide map analyses by LC–MS
played an important role for the characterization of the EtO–
methionine modifications in Peg-GCSF and HSA in this study.
We have closely examined the degradation of these proteins
in different EtO sterilized containers and found a characteris-
tic MS/MS fragmentation pattern for the methionine-modified
peptides. Although the modified peptide showed a mass in-
crease of 44 Da, its fragmentation mass spectra (MS/MS) often
contained a peak where the mass decreased by 92 Da relative to
the precursor ion. It appeared that the side chain of the methio-
nine residue was increased by 44 Da and was then lost during
the gas-phase fragmentation generating a total loss of 92 Da
relative to the modified peptide, which resulted in a net change
of 48 Da. We have found that an EtO-modified methionine side
chain has a unique fragmentation pattern under CID in that it
readily loses a C3H8OS group (92 Da). The loss of the side chain
and double-cleavage (side chain and peptide backbone) create
a fragmentation pattern that is difficult to identify for typical
software algorithms. The uncovered gas-phase fragmentation
pattern of peptides with methionine residues modified by EtO
should facilitate reliable and automated identification in future
analyses of this modification by LC–MS/MS in therapeutic pro-
teins.
Regarding the other biophysical and biochemical properties
of EtO adducts, the EtO modification of methionine residues in-
volved formation of a positively charged sulfonium compound
resulting from EtO reacting with the sulfur ion of methionine.
Windmueller et al.1reported in a model experiment that sul-
fonium derivatives of methionine possess different crystalliza-
tion ability and water solubility. In the present study, the EtO–
methionine modification species (EtO–methionine adducts) in
Peg-GCSF were eluted at the post peaks region on CEX–HPLC.
It was also found eluting in the prepeak region with less hy-
drophobicity than the oxidized methionine species on RP-HPLC
analysis of Peg-GCSF and HSA.
For the EtO-cysteine reaction, MS/MS of peptides containing
a modified cysteine were detected by tryptic peptide mapping
of the HSA. The modified peptide with a 14 Da difference
compared to the native peptide (A21-K41) was identified as
an EtO–cysteine modification. The 14 Da was the result of a
mass difference between alkylated-cysteine (+58 Da) and EtO-
cysteine (+44 Da).
Ethylene oxide modifications can alter the stability and
bioactivity of proteins and several published reports15 have
shown the impact of EtO-modified proteins on patient safety.
The possibility of EtO modifications of therapeutic proteins and
the potential impact should be considered during EtO steriliza-
tion processes development. Additional methods to confirm that
residual EtO have reached acceptable levels and the applica-
tion of these methods to understand the desorption kinetics of
EtO in a variety of materials would be advantageous for future
drug and container development.
ACKNOWLEDGMENTS
The authors would like to thank Gang Xiao for fruitful discus-
sions and support in performing LC–MS analyses, and Mike
Treuheit for careful review of the manuscript and valuable
suggestions.
REFERENCES
1. Windmueller HG, Ackerman CJ, Engel RW. 1958. Reaction of ethy-
lene oxide with histidine, methionine and cysteine. J Biol Chem
234(4):895–899.
2. Kelsey JC. 1961. Sterilization by ethylene oxide. J Clin Pathol 14:59–
61.
3. Ford DJ. 1976. The effect of methods of sterilization on the nu-
tritive value of protein in a commercial rat diet. Br J Nutr 35:267–
276.
4. Wu KY, Chiang SY, Shih WC, Huang CC, Chen MF, Swenberg
JA. 2011. The application of mass spectrometry in molecular dosime-
try: Ethylene oxide as an example. Mass Spectrom Rev 30(5):733–
756.
5. Mendes GCC, Brandao TRS, Silva CLM. 2007. Ethylene oxide ster-
ilization of medical devices: A review. Am J Infect Control 35:574–
581.
6. Nakata S, Umeshita K, Ueyama H, Takashina M, Noguchi S, Mu-
rata A, Ochi T. 2000. Aeration time following ethylene oxide ster-
ilization for reusable rigid sterilization containers: Concentration of
gaseous ethylene oxide in containers. Biomed Instrum Technol 34:121–
124.
7. Lucas AD, Merritt K, Hitchins VM, Woods TO, McNamee SG, Lyle
DB, Brown SA. 2003. Residual ethylene oxide in medical devices and
device material. J Biomed Mater Res Part B 66B:548–552.
8. Buben L, Melichercikova V, Novotna N, Svitakova R. 1999. Problems
associated with sterilization using ethylene oxide residues in treated
materials. Centr Eur J Publ Hlth 7(4):197–202.
9. Dias FN, Ishii M, Nogaroto Sl, Piccini B, Penna TCV. 2009. Ster-
ilization of medical devices by ethylene oxide, determination of the
dissipation of residues, and use of green fluorescent protein as an
indicator of process control. J Biomed Mater Res Part B 91B:626–
630.
10. Pan B, Abel J, Ricci M, Brems D, Wang D, Trout B. 2006. Com-
parative oxidation studies of methionine residues reflect a struc-
tural effect on chemical kinetics in rhG-CSF. Biochemistry 45:15430–
15443.
11. Chu J, Yin J, Wang D, Trout B. 2004. Molecular dynamics sim-
ulations and oxidation rates of methionine residues of Granulocyte
Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24257
RESEARCH ARTICLE Pharmaceutics, Drug Delivery and Pharmaceutical Technology 9
colony-stimulating factor at different pH values. Biochemistry
43:1019–1029.
12. Jiang X, Smith JB, Abraham EC. 1996. Identification of a MS–
MS fragment diagnostic for methionine sulfoxide. J Mass Spectrom
31(11):1309–1310.
13. Zhang Z. 2011. Prediction of collision-induced-dissociation spectra
of peptides with post-translational or process-induced modifications.
Anal Chem 83:8642–8651.
14. Beck JL, Ambahera S, Yong SR, Sheil MM, de Jersey J, Ralph
SF. 2004. Direct observation of covalent adducts with Cys34 of human
serum albumin using mass spectrometry. Anal Biochem 325(2):326–
336.
15. Grammer LC, Patterson R. 1987. IgE against ethylene-altered
human serum albumin (EtO-HSA) as an etiologic agent in al-
lergic reactions of hemodialysis patients. Artif Organs 11:97–
99.
DOI 10.1002/jps.24257 Chen et al., JOURNAL OF PHARMACEUTICAL SCIENCES
... Studies have shown that sterilization of prefilled syringes with ethylene oxide (EO) can lead to methionine, cysteine, and histidine EO adducts. 21,22 To assess the influence of EO during the filling of a bispecific antibody (BsMAb), forced degradation studies were performed. For the identification of susceptible amino acids, the samples were analyzed by mD-UPLC-MS, and the results were used for further process optimization. ...
Article
Full-text available
For the past few years, multidimensional liquid chromatography-mass spectrometry (LC-MS) systems have been commonly used to characterize post-translational modifications (PTMs) of therapeutic antibodies (mAbs). In most cases, this is performed by fractionation of charge variants by ion-exchange chromatography and subsequent online LC-MS peptide mapping analysis. In this study, we developed a multidimensional ultra-performance-liquid-chromatography-mass spectrometry system (mD-UPLC-MS/MS) for PTM characterization and quantification, allowing both rapid analysis and decreased risk of artificial modifications during sample preparation. We implemented UPLC columns for peptide mapping analysis, facilitating the linkage between mD-LC and routine LC-MS workflows. Furthermore, the introduced system incorporates a novel in-parallel trypsin and LysC on-column digestion setup, followed by a combined peptide mapping analysis. This parallel digestion with different enzymes enhances characterization by generating two distinct peptides. Using this approach, a low retentive ethylene oxide adduct of a bispecific antibody was successfully characterized within this study. In summary, our approach allows versatile and rapid analysis of PTMs, enabling efficient characterization of therapeutic molecules.
... 30 The stability of therapeutic protein formulations in EtO-sterilized plastic vials was investigated and the authors found that residual EtO rapidly formed adducts with methionine residues in pegylated granulocyte colony stimulating factor and caused similar chemical modifications to human serum albumin (HSA). 31 Based on other studies, these types of changes in the protein CQAs could potentially have adverse effects in patients. For example, it was found that EtO adducts of HSA that were formed in dialysis machines at residual EtO levels below the limits set by the International Organization for Standardization could cause anaphylaxis. ...
Article
Protein-based biologic drugs encounter a variety of stress factors during drug substance (DS) and drug product (DP) manufacturing, and the subsequent steps that result in clinical administration by the end user. This article is the third in a series of commentaries on these stress factors and their effect on biotherapeutics. It focuses on assessing the potential negative impact from primary packaging, transportation, and handling on the quality of the DP. The risk factors include ingress of hazardous materials such as oxidizing residuals from the sterilization process, delamination- or rubber stopper-derived particles, silicone oil droplets, and leachables into the formulation, as well as surface interactions between the protein and packaging materials, all of which may cause protein degradation. The type of primary packaging container used (such as vials and prefilled syringes) may substantially influence the impact of transportation and handling stresses on DP Critical Quality Attributes (CQAs). Mitigations via process development and robustness studies as well as control strategies for DP CQAs are discussed, along with current industry best practices for scale-down and in-use stability studies. We conclude that more research is needed on postproduction transportation and handling practices and their implications for protein DP quality.
... Furthermore, oxidized proteins were prone to aggregation during the lyophilization process (64). Similar to vaporized hydrogen peroxide, detrimental effects of ethylene oxide have been reported in the literature; significant degradation of human serum albumin and pegylated granulocyte-colony stimulating factor have been reported (66). Therefore, undesired ingress of ethylene oxide into the drug product has been demonstrated to be detrimental to the drug product depending on protein oxidation potential, formulation characteristics, and primary container material, all of which have the potential to for unwanted clinical consequences (67). ...
Article
Full-text available
Intravitreal (IVT) administration of therapeutics is the standard of care for treatment of back-of-eye disorders. Although a common procedure performed by retinal specialists, IVT administration is associated with unique challenges related to drug product, device and the procedure, which may result in adverse events. Container closure configuration plays a crucial role in maintaining product stability, safety, and efficacy for the intended shelf-life. Careful design of primary container configuration is also important to accurately deliver small volumes (10-100 μL). Over- or under-dosing may lead to undesired adverse events or lack of efficacy resulting in unpredictable and variable clinical responses. IVT drug products have been traditionally presented in glass vials. However, pre-filled syringes offer a more convenient administration option by reducing the number of steps required for dose preparation there by potentially reducing the time demand on the healthcare providers. In addition to primary container selection, product development studies should focus on, among other things, primary container component characterization, material compatibility with the formulation, formulation stability, fill volume determination, extractables/leachables, and terminal sterilization. Ancillary components such as disposable syringes and needles must be carefully selected, and a detailed administration procedure that includes dosing instructions is required to ensure successful administration of the product. Despite significant efforts in improving the drug product and administration procedures, ocular safety concerns such as endophthalmitis, increased intraocular pressure, and presence of silicone floaters have been reported. A systematic review of available literature on container closure and devices for IVT administration can help guide successful product development.
Article
Full-text available
Titanium implants are widely used in medicine but have a risk of biomaterial‐associated infection (BAI), of which traditional antibiotic‐based treatment is affected by resistance. Antimicrobial peptides (AMPs) are used to successfully kill antibiotic‐resistant bacteria. Herein, a supramolecular coating for titanium implants is developed which presents the synthetic antimicrobial and antibiofilm peptide SAAP‐148 via supramolecular interactions using ureido‐pyrimidinone supramolecular units (UPy‐SAAP‐148GG). Material characterization of dropcast coatings shows the presence of UPy‐SAAP‐148GG at the surface. The supramolecular immobilized peptide remains antimicrobially active in dropcast polymer films and can successfully kill (antibiotic‐resistant) Staphylococcus aureus, Acinetobacter baumannii, and Escherichia coli. Minor toxicity for human dermal fibroblasts is observed, with a reduced cell attachment after 24 h. Subsequently, a dipcoat coating on titanium implants is developed and tested in vivo in a subcutaneous implant infection mouse model with S. aureus administered locally on the implant before implantation to mimic contamination during surgery. The supramolecular coating containing 5 mol% of UPy‐SAAP‐148GG significantly prevents colonization of the implant surface as well as of the surrounding tissue, with no signs of toxicity. This shows that supramolecular AMP coatings on titanium are eminently suitable to prevent BAI.
Article
Many pharmaceutical manufacturing units utilize pre-sterilized ready-to fill primary containers for parenterals. The containers may have been sterilized by the supplier via autoclavation. This process can change the physicochemical properties of the material and the subsequent product stability. We studied the impact of autoclavation on baked on siliconized glass containers for biopharmaceuticals. We characterized the container layers of different thickness before and after autoclavation for 15 min at 121 °C and 130 °C. Furthermore, we analyzed the adsorption of a mAb to the silicone layer and subjected filled containers to 12 weeks storage at 40 °C monitoring functionality and subvisible particle formation of the product. Autoclavation turned the initially homogenous silicone coating into an incoherent surface with uneven microstructure, changed surface roughness and energy, and increased protein adsorption. The effect was more pronounced at higher sterilization temperatures. We did not observe an effect of autoclavation on stability. Our results did not indicate any concerns for autoclavation at 121 °C for safety and stability of drug/device combination products using baked-on siliconized glass containers.
Article
Degradable and environmentally responsive polymers have been actively developed for drug delivery and regenerative medicine applications, yet inadequate consideration of their compatibility with terminal sterilization presents notable barriers to clinical translation. This Review discusses industry-established terminal sterilization methods and aseptic processing and contrasts them with innovative approaches aimed at preserving the integrity of polymeric implants. Regulatory guidelines, fiscal considerations, and potential pitfalls are discussed to encourage early integration of sterility regulatory considerations in material designs.
Article
Full-text available
Recent advances in the field of biomedical materials have demonstrated that covalent mucin coatings generated on polymeric materials have the potential to greatly improve the surface properties of medical devices such as their wettability, lubricity, and resistance toward biofouling. For such biopolymer‐based coatings to be used in a medical application, sterilization of the coated devices is mandatory. However, common sterilization methods such as autoclavation, ethylene oxide fumigation, as well as γ‐ or ultraviolet‐irradiation, create harsh conditions during the device treatment, and this might compromise the structural integrity and thus functionality of macromolecular coatings. Here, it is demonstrated that covalent mucin coatings generated on medical devices made from polyvinyl chloride, polyurethane, or polydimethylsiloxane are able to withstand such treatments—albeit to different extents. Among all treatments tested, ethylene oxide fumigation is identified as the most promising method as it maintains the coatings the best. The findings imply that the beneficial properties demonstrated for mucin coatings in vitro should indeed be transferable to applications in vivo. Covalent mucin coatings can improve the wettability, lubricity, and anti‐biofouling behavior of polymeric surfaces. The influence of γ irradiation, autoclaving, ethylene oxide, or ultraviolet irradiation on such coatings generated on medical devices made from polyvinyl chloride (endotracheal tube), polyurethane (urinary catheter), or polydimethylsiloxane (contact lens) is investigated.
Article
Rapid technological advances have significantly improved the capability, versatility, and robustness of mass spectrometers which has led to them playing a central role in the development, characterization, and regulatory filings of biopharmaceuticals. Their application spans the entire continuum of drug development, starting with discovery research through product development, characterization, and marketing authorization and continues well into product life cycle management. The scope of application extends beyond traditional protein characterization and includes elements like clone selection, cell culture physiology and bioprocess optimization, investigation support, and process analytical technology. More recently, advances in the MS-based multi-attribute method are enabling the introduction of MS in a cGMP environment for routine release and stability testing. While most applications of MS to date have been for monoclonal antibodies, the successes and learnings should translate to the characterization of next-gen biotherapeutics where modalities like multispecifics could be more prevalent. In this review, we describe the most significant advances in MS and correlate them to the broad spectrum of applications to biotherapeutic development. We anticipate rapid technological improvements to continue that will further accelerate widespread deployment of MS, thereby elevating our overall understanding of product quality and enabling attribute-focused product development.
Article
To reduce the risk of infection during intravitreal injections, the external surface of prefilled syringes (PFSs) must be sterilized. Usually, ethylene oxide (EO) gas or vaporized hydrogen peroxide (VHP) is used for sterilization. More recently, nitrogen dioxide (NO2) gas sterilization has been developed. It is known that gas permeability is approximately zero into glass-PFSs. However, polymer-PFSs (P-PFSs) have relatively high gas permeability. Therefore, there are concerns about the potential impact of external surface sterilization on drug solutions in P-PFSs. In this study, P-PFSs [filled with water for injection (WFI) or human serum albumin (HSA) solution] were externally sterilized using EO, VHP, and NO2 gases. For the WFI-filled syringes, the concentration of each gas that ingressed into the WFI was measured. For the HSA solution-filled syringes, the physical and chemical degradation of HSA molecules by each sterilant gas was quantified. For the EO- or VHP-sterilized syringes, the ingressed EO or hydrogen peroxide (H2O2) molecules were detected in the filled WFI. Additionally, EO-adducted or oxidized HSA molecules were observed in the HSA-filled syringes. In contrast, the NO2-sterilized WFI-filled syringes exhibited essentially immeasurable ingressed NO2, and protein degradation was not detected in HSA-filled syringes.
Article
Full-text available
Mucin glycoproteins, the macromolecular components of mucus, combine a broad range of biomedically important properties. Among those is the ability of mucin solutions to act as excellent lubricants. However, to be able to use purified, endogenous mucin glycoproteins as components of a biomedical product, the mucins need to be sterile; this, in turn, makes it necessary to subject the mucins to quite harsh physical treatments, such as heat exposure, autoclaving, UV‐, or γ‐irradiation, which might compromise the functionality of the glycoproteins. Here, it is shown that mucins are indeed able to withstand most of those treatments without suffering significant lubrication impairment or structural degradation. Among those treatments, which left the mucins unharmed, γ‐irradiation is identified to be the most powerful one in terms of inactivating microbial contaminations. The obtained results demonstrate a remarkable sturdiness of mucins, which opens up broad possibilities for them to be further processed into materials, e.g., as parts of biomedical products.
Article
Full-text available
Ethylene oxide (EO) gas is commonly used to sterilize medical devices. The amount of residual EO remaining in a device depends partly on the type and size of polymeric material. A major concern is the amount of residue that may be available in the body. With the use of the method described by AAMI for headspace analysis of EO residues, different polymers and medical devices subjected to different numbers of sterilization cycles were examined. Next, the effect of various extraction conditions and extraction solutions on these polymers and medical devices was evaluated. The results showed different polymers desorb EO differently. One polyurethane (PU 75D) had much higher EO residue than a different polyurethane (PU 80A). Repeated extraction of the PU 75D was necessary to quantify total EO residue levels. Different extraction solutions influence the amount and reproducibility of EO detected, whereas multiple resterilizations showed no difference in amount of residual EO. Bioavailability of EO was estimated by extracting the devices and polymers in water. Comparison of total EO residues to EO that was bioavailable showed no difference for some polymers and devices, while others had an almost eightfold difference. Some standard biocompatibility tests were run on extracts and devices, but no significant effects were observed.
Article
Ethylene oxide (ETO) is used to sterilize hemodialyzers and other medical equipment. In an attempt to confirm a link between ETO and hypersensitivity reactions during hemodialysis (HD) we quantitated IgE and total antibody directed against ETO-altered human serum albumin (ETO-HSA) in the sera of 65 hemodialysis patients. In 24 patients who had experienced anaphylaxis during HD. the levels of IgE and total antibody against ETO-HSA were significantly higher than the corresponding levels in 41 patients who had not. Our data demonstrate an association between the presence of IgE and total antibody against ETO-HSA and immediate anaphylactic reactions to HD. In further studies we characterized the ETO-HSA antigen by Immunoelectrophoresis, gel filtration chromatography, and cross-inhibition immunoassay. Our results suggested that ETO gas can alter HSA and induce new antigenic determinants on the molecule. Recently, we encountered a peritoneal dialysis patient with rash and eosinophilia. Suspecting ETO allergy, we measured serum IgE and total antibody to ETO-HSA and found both to be present. The data suggest that, in addition to the familiar HD reactions, ETO sensitization might cause other allergic diseases as well.
Article
Mass spectrometry, combined with collision-induced dissociation (CID), has become the method of choice for analyzing protein post-translational and process-induced modifications. However, confident and automated identification of modifications and modification sites is often challenged by the diversity of modifications and their labile nature under typical CID conditions. An accurate prediction of the CID spectra of modified peptides will improve the reliability of automated determination of modifications and modification sites. In this article, the kinetic model for the prediction of peptide CID spectra is extended to the prediction of the CID spectra of modified peptides. The mathematical model for predicting CID spectra of peptides with enzymatic and chemical modifications such as (1) phosphorylation of serine, threonine, and tyrosine, (2) S-carboxymethylation and carbamidomethylation of cysteine, (3) different stages of oxidation of methionine, tryptophan, and cysteine, (4) glycation of lysine, (5) O-mannosylation of serine, (6) hydroxylation of lysine, and (7) N-monomethylation and N-dimethylation of lysine is described. The mathematical model, once established with CID spectra of peptides with known modifications and modification sites, is able to predict CID spectra with excellent accuracy in ion intensities, facilitating more reliable identification of modification and modification sites.
Article
Mass spectrometry plays an increasingly important role in the search for and quantification of novel chemically specific biomarkers. The revolutionary advances in mass spectrometry instrumentation and technology empower scientists to specifically analyze DNA and protein adducts, considered as molecular dosimeters, derived from reactions of a carcinogen or its active metabolites with DNA or protein. Analysis of the adducted DNA bases and proteins can elucidate the chemically reactive species of carcinogens in humans and can serve as risk-associated biomarkers for early prediction of cancer risk. In this article, we review and compare the specificity, sensitivity, resolution, and ease-of-use of mass spectrometry methods developed to analyze ethylene oxide (EO)-induced DNA and protein adducts, particularly N7-(2-hydroxyethyl)guanine (N7-HEG) and N-(2-hydroxyethyl)valine (HEV), in human samples and in animal tissues. GC/ECNCI-MS analysis after HPLC cleanup is the most sensitive method for quantification of N7-HEG, but limited by the tedious sample preparation procedures. Excellent sensitivity and specificity in analysis of N7-HEG can be achieved by LC/MS/MS analysis if the mobile phase, the inlet (split or splitless), and the collision energy are properly optimized. GC/ECNCI-HRMS and GC/ECNCI-MS/MS analysis of HEV achieves the best performance as compared with GC/ECNCI-MS and GC/EI-MS. In conclusion, future improvements in high-throughput capabilities, detection sensitivity, and resolution of mass spectrometry will attract more scientists to identify and/or quantify novel molecular dosimeters or profiles of these biomarkers in toxicological and/or epidemiological studies. © 2011 Wiley Periodicals, Inc. Mass Spec Rev.
Article
Ethylene oxide (EO) is used to sterilize Oxygenator and Tubing applied to heart surgery. Residual levels of EO and its derivatives, ethylene chlorohydrin (ECH) and ethylene glycol (EG), may be hazardous to the patients. Therefore, it must be removed by the aeration process. This study aimed to estimate the minimum aeration time for these devices to attain safe limits for use (avoiding excessive aeration time) and to evaluate the Green Fluorescent Protein (GFP) as a biosensor capable of best indicating the distribution and penetration of EO gas throughout the sterilization chamber. Sterilization cycles of 2, 4, and 8 h were monitored by Bacillus atrophaeus ATCC 9372 as a biological indicator (BI) and by the GFP. Residual levels of EO, ECH, and EG were determined by gas chromatography (GC), and the residual dissipation was studied. Safe limits were reached right after the sterilization process for Oxygenator and after 204 h of aeration for Tubing. In the 2 h cycle, the GFP concentration decreased from 4.8 (+/-3.2)% to 7.5 (+/-2.5)%. For the 4 h cycle, the GFP concentration decreased from 17.4 (+/-3.0)% to 21.5 (+/-6.8)%, and in the 8 h cycle, it decreased from 22.5 (+/-3.2)% to 23.9 (+/-3.9)%. This finding showed the potentiality for GFP applications as an EO biosensor.
Article
1. The effect on protein quality of treating a commercial rat diet by autoclaving at various temperatures for different periods of time, or by irradiation with 2.5 or 10 Mrd, was studied. True digestibility (TD) and biological value (BV) were measured and the available and total amino acids in the diets were estimated using microbiological and chemical methods.. 2. Autoclaving at 121° for 60 min reduced BV, TD and net protein utilization (NPU) more than autoclaving at 134° for 3 min. Availability of amino acids was reduced by both treatments but to a greater extent by autoclaving at 121° for 60 min. Total amino acids were essentially unaffected. Irradiation had no effect on BV, TD, NPU or total amino acids, and the availability of amino acids was also unaffected, with the exception of lysine which was slightly reduced.. 3. When the diet was autoclaved at 115 or 121° for 15, 30 or 60 min, or at 134° for 3 min the availability of the amino acids was reduced with increasing time and temperature of treatment. Treatment at 134° for 3 min had an effect on available amino acids similar to treatment at 121° for 15 or 30 min.. 4. Ethylene oxide fumigation of the diet caused reduced availability of histidine, methionine and tryptophan but had negligible effect on arginine, leucine and lysine.. 5. It is concluded that from a practical point of view irradiation causes least damage to proteins in rodent diets. If such diets are to be autoclaved they should be supplemented with complete protein to counteract amino acid destruction.
Article
The paper deals with problems associated with reduction of undesirable effects of ethylene oxide in polymers in medical devices on the patient's health. The authors explain the need of careful elaboration and validation of the sterilization and aeration process incl assessment of ethylene oxide (EO) residues. The authors investigated the effect of the type of material and conditions of sterilization and aeration on the assessed EO concentration. For research of the behaviour of different polymers in the sterilization process model sterilizations of actual items of medical devices with a known composition proved more suitable than assessment in medical devices from medical institutions. The main conclusions of the investigation were a classification of polymers into those suitable and unsuitable for sterilization or resterilization, and attention was also drawn to poor reproducibility of results in old sterilizers, in particular those lacking effective aeration in aerators.
Article
Because ethylene oxide (EO) gas is toxic to humans, restrictions have been imposed on its use for sterilization, specifying allowable levels of residual EO remaining in sterilized apparatus and materials. However, the aeration time that optimizes the removal of the remaining EO when a rigid sterilizing container is used for a vessel had not been identified. Therefore, polyvinyl chloride, which easily adsorbs EO, was placed in rigid sterilizing containers, and aeration was carried out after 1, 8, 12, 17, and 24 hours. After standard EO sterilization, the EO concentrations remaining in the air in the rigid containers were measured. The results indicate that a period of 17 hours of aeration is appropriate when a rigid sterilizing container is used.