Accepted Manuscript Bioavailability of protein therapeutics in rats following inhalation exposure: Relevance to occupational exposure limit calculations Janet C. Gould, Irvith Carvajal, Todd Davidson, Jessica Graham, Jedd Hillegass, Susan Julien, Alex Kozhich, Bonnie Wang, Hui Wei, Aaron P. Yamniuk, Neil Mathias, Helen G. Haggerty, Michael Graziano PII:
S0273-2300(18)30254-X
DOI:
10.1016/j.yrtph.2018.10.003
Reference:
YRTPH 4233
To appear in:
Regulatory Toxicology and Pharmacology
Received Date: 16 April 2018 Revised Date:
27 September 2018
Accepted Date: 2 October 2018
Please cite this article as: Gould, J.C., Carvajal, I., Davidson, T., Graham, J., Hillegass, J., Julien, S., Kozhich, A., Wang, B., Wei, H., Yamniuk, A.P., Mathias, N., Haggerty, H.G., Graziano, M., Bioavailability of protein therapeutics in rats following inhalation exposure: Relevance to occupational exposure limit calculations, Regulatory Toxicology and Pharmacology (2018), doi: https://doi.org/10.1016/ j.yrtph.2018.10.003. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
1
Bioavailability of protein therapeutics in rats following inhalation exposure: relevance to
2
occupational exposure limit calculations
RI PT
3 Janet C. Goulda, Irvith Carvajalb, Todd Davidsonb, Jessica Grahamb, Jedd Hillegassb*, Susan Julienb, Alex
5
Kozhichb, Bonnie Wangb, Hui Weib, Aaron P. Yamniukb, Neil Mathiasb, Helen G. Haggertyb, Michael
6
Grazianob
SC
4
7 a
SafeBridge Consultants, Inc., 330 Seventh Avenue – Suite 1101, New York, NY 10001, USA
9
b
Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, NJ 08903, USA
M AN U
8
10 *Corresponding author:
12
Jedd Hillegass, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, NJ 08903, USA
13
[email protected]; 732-227-6093
16 17 18
Word count: Abstract (200); Text (7712); References (978)
EP
15
AC C
14
TE D
11
19 20 21 1
ACCEPTED MANUSCRIPT
Abstract
23
Protein therapeutics represent a rapidly growing proportion of new medicines being developed by the
24
pharmaceutical industry. As with any new drug, an Occupational Exposure Limit (OEL) should be
25
developed to ensure worker safety. Part of the OEL determination addresses bioavailability (BA) after
26
inhalation, which is poorly understood for protein therapeutics. To explore this, male Sprague-Dawley
27
rats were exposed intravenously or by nose-only inhalation to one of five test proteins of varying
28
molecular size (10-150 kDa), including a polyethylene glycol-conjugated protein. Blood, lung tissue and
29
bronchoalveolar lavage (BAL) fluid were collected over various time-points depending on the expected
30
test protein clearance (8 minutes-56 days), and analyzed to determine the pharmacokinetic profiles. Since
31
the BAL half-life of the test proteins was observed to be >4.5 hours after an inhalation exposure,
32
accumulation and direct lung effects should be considered in the hazard assessment for protein
33
therapeutics with lung-specific targets.
34
inhalation exposure for all test proteins (~≤1%) which did not appear molecular weight-dependent. Given
35
that this study examined the inhalation of typical protein therapeutics in a manner mimicking worker
36
exposure, a default 1% BA assumption is reasonable to utilize when calculating OELs for protein
37
therapeutics.
M AN U
SC
RI PT
22
TE D
The key finding was the low systemic bioavailability after
EP
38
Key words: Inhalation, Protein Therapeutic, Antibody, Pharmacokinetics, Occupational Exposure Limit,
40
Bioavailability
41
AC C
39
42
Abbreviations:
43
AUC – area under the curve
44
BA – bioavailability
45
BAL – bronchoalveolar lavage
46
BSA – bovine serum albumin
47
BTLA mAb – B and T lymphocyte attenuator human monoclonal IgG1antibody 2
ACCEPTED MANUSCRIPT
Cmax – maximum serum concentration
49
Da – dalton
50
DNA – deoxyribonucleic acid
51
EDC – ethyl(dimethylaminopropyl) carbodiimide
52
Fc – crystallization fragment
53
FcRn – neonatal constant region fragment receptor
54
FDA – Food and Drug Administration
55
GSD – Geometric standard deviation
56
IgG – immunoglobulin G
57
IL23 – interleukin 23
58
IL23 scFv – single chain fragment variable against IL-23
59
HPLC – high performance liquid chromatography
60
HRP – horseradish peroxidase
61
IV – intravenous
62
KD – dissociation constant
63
kDa – kilodalton
64
λz – elimination rate constant
65
LLOQ – lower limit of quantitation
66
LOAEL – lowest observed adverse effect level
67
µg – microgram
68
µm – micrometer
69
mg – milligram
70
MMAD – mass median aerodynamic diameter
71
MSD – meso scale discovery
72
MW – molecular weight
73
NBCA– non-binding control adnectin
AC C
EP
TE D
M AN U
SC
RI PT
48
3
ACCEPTED MANUSCRIPT
NBCA-PEG – pegylated non-binding control adnectin
75
nm – nanometer
76
NIOSH – National Institute of Occupational Safety and Health
77
NOAEL – no observed adverse effect level
78
OEL – occupational exposure limit
79
PBS – phosphate buffered saline
80
PoD – point of departure
81
PEG – polyethylene glycol
82
pI – isoelectric point
83
PK – pharmacokinetic
84
PNOS – particles not otherwise specified
85
RNA – ribonucleic acid
86
SPR – surface plasmon resonance
87
T1/2 – terminal half-life
88
Tmax – time to maximum concentration
89
UPLC/MS – ultra performance liquid chromatography/mass spectrometry
92 93 94 95
SC M AN U
TE D
EP
91
AC C
90
RI PT
74
96 97 98 99 4
ACCEPTED MANUSCRIPT
100 1. Introduction
102
Biologics are an increasing part of the pharmaceutical market today, with Biologic License Applications
103
making up approximately one-third of the new drugs approved within the last two years by the Food and
104
Drug Administration (FDA) [1]. Technological advances in peptide and protein engineering have led to
105
the emergence of a range of novel constructs, which can vary greatly in size and complexity. Monoclonal
106
antibodies (~150 kDa), fusion proteins, protein and antibody fragments, and analogs to small proteins or
107
peptides (<5 kDa) represent a significant component of the biological drug landscape. The broad
108
description of a biologic is any material produced by a natural source such as an endogenous protein or
109
any substance derived from bacterial or mammalian cells [2]. Their typical manufacture involves large-
110
scale cell culture, filtration and purification of the protein. While processing requires protection of
111
product, individual workers may still come in contact with the biologic drug through aerosol generation in
112
positive pressure systems or in upset conditions. In addition, pharmacists and healthcare providers handle
113
the finished drug to prepare and administer it to patients. Thus, there is a need to understand the hazard
114
potential and develop occupational exposure limits (OEL) to establish appropriate exposure controls for
115
the workplace.
116
An OEL is an air concentration to which workers can be exposed for an 8-hour day, 5 days a week for a
117
40-year career protecting them from experiencing pharmacological or toxicological effects of the
118
substance. In order to assign an OEL, the preclinical and clinical safety data and pharmacokinetic (PK)
119
properties are evaluated for each protein therapeutic. Though other approaches exist, an OEL is most
120
commonly calculated by the following formula: OEL = PoD x α / FT x AF x V, where PoD is the point of
121
departure (usually a No-Observed Adverse-Effect Level [NOAEL] or a Lowest-Observed Adverse Effect
122
Level [LOAEL] for the critical effect); α is an adjustment factor to correct for differences in
123
bioavailability (BA) between the route of administration from the study from which the PoD was selected
124
and the inhalation route; FT is the composite adjustment factor which may account for 1) interspecies
125
variation, 2) intraspecies variation, 3) study duration, 4) severity of the effect and database completeness,
AC C
EP
TE D
M AN U
SC
RI PT
101
5
ACCEPTED MANUSCRIPT
and 5) LOAEL to NOAEL extrapolation; AF is a factor used to account for potential accumulation
127
following repeated exposure; and V is the breathing volume in an 8-hour workday. Additional details on
128
these factors and how they are applied in OEL estimation can be found in several publications [3-5]. The
129
research provided in this manuscript focuses on generation of α for protein therapeutics following
130
inhalation exposure.
131
Protein therapeutics are generally thought to be less hazardous for workers than traditional small molecule
132
drugs because of their likeness to intrinsic human molecules (i.e., higher specificity, susceptibility to
133
enzymatic degradation to amino acids, with minimal nonspecific toxicity), expected instability at room
134
temperature, and negligible dermal bioavailability (generally considered to be poorly absorbed due to
135
their large size); factors combined with the limited opportunity for inhalation (e.g., present as a dilute
136
solution in primarily closed systems). However, since proteins can be systemically absorbed to some
137
degree by the inhalation route [6-11], a more accurate assessment of their BA should be considered when
138
assessing potential health effects in workers and in establishing OEL values. Since data on the inhalation
139
BA of protein therapeutics are generally not available, most OELs for these substances are derived from
140
clinical data using intravenous (IV) or subcutaneous injections and extrapolating to an inhalation BA of
141
5% primarily based on studies in various species utilizing intratracheal dosing [6] or greater than 5%
142
based on the NIOSH Hazardous Drug Alert List [12]. As a consequence, OELs for protein therapeutics
143
likely over-predict the systemic presence of the drug following inhalation exposure, which can then lead
144
to the implementation of overly conservative and potentially unnecessary environmental controls for
145
workers. In addition, most of the published data on BA of protein therapeutics was developed using
146
intratracheal delivery, specialized inhalation devices, and/or formulations.
147
designed to enhance systemic drug delivery via the inhalation route; none of which accurately reflect
148
normal breathing of an aerosolized protein in the workplace. Finally, whereas there are some reports that
149
describe the inhalation delivery of peptide hormones like insulin (~6 kDa) and antibodies (~150 kDa), the
150
BA of novel therapeutics that lie in a range of intermediate sizes (~10-150 kDa) is highly varied and
AC C
EP
TE D
M AN U
SC
RI PT
126
6
These technologies are
ACCEPTED MANUSCRIPT
incomplete. Therefore, a critical question exists as to the most appropriate inhalation BA adjustment
152
factor to be utilized for calculating an OEL for these types of molecules.
153
In order to identify a default BA factor(s), the test proteins in the current study were selected to mimic
154
typical protein therapeutics. Besides the molecular weight (MW) or size, the binding properties at the
155
therapeutic target in the lung was considered in selection of the test proteins. Two protein features (i.e.,
156
crystallization fragment [Fc] region present in Immunoglobulin G [IgG] and albumin) or modifications
157
(i.e., polyethylene glycol [PEG] conjugation) and their influence on pharmacokinetics were examined.
158
The neonatal constant region fragment receptor (FcRn) facilitates IgG crossing of epithelial barriers (e.g.,
159
placenta, neonatal intestinal epithelium, and adult lung) by receptor-mediated transcytosis with reduced
160
lysosomal degradation [13, 14] leading to increased systemic absorption and circulating half-life after
161
dosing to the lung [15, 16]. Likewise, protein PEGylation with its hygroscopic nature, increasing the
162
overall molecule’s apparent size, has been shown to influence respiratory tract pharmacokinetics [17, 18].
163
In essence, these factors were considered in selecting the protein therapeutics for the study.
164
The work presented in this manuscript aims to provide information on BA for typical protein therapeutics
165
with increasing molecular weight that can serve as a model to understand human lung and systemic
166
exposure to an aerosolized drug solution. The application of these data to human inhalation BA estimates
167
and its impact on the calculation of OELs will be discussed. Additionally, underlying transport properties
168
were examined, as these relate to the lung absorption and disposition of different types of protein
169
therapeutics. The potential for local effects following inhalation of proteins (e.g., specific binding to lung
170
tissue, target receptor lung effects) are not addressed in this paper.
SC
M AN U
TE D
EP
AC C
171
RI PT
151
172
2. Materials and Methods
173
2.1. Materials
174
NBCA, IL23 scFv, BTLA mAb and NCBA-PEG were synthesized, purified and verified in the Bristol-
175
Myers Squibb laboratories. Description of the test proteins, their molecular weight and isoelectric point
7
ACCEPTED MANUSCRIPT
176
(pI) are listed in Table 1. BSA (essentially fatty acid and globulin free) was purchased from Sigma-
177
Aldrich.
178
RI PT
179 180
Table 1
183
Description of test proteins. Test Protein
Molecular Weight (kDa) 10.9
Isoelectric Pointb (pI) 5.14
FcRn binding c KD (µM) NA
M AN U
182
SC
181
NBCA
Non-binding control adnectina
IL23 scFv
Anti-interleukin 23 single-chain variable fragment
30.1
6.18
NA
BSA
Bovine serum albumin
67
5.40
Human = 3.7 Monkey = 2.2 Rat = NDBe Mouse = NDB
BTLA mAb (8A3)
Anti-B and T lymphocyte attenuator -human monoclonal IgG1antibody
144
Heavy chain: 9.08 Light chain: 7.60 Intact mAb: 8.90
Human = 3.0 Monkey = 2.4 Rat = 1.2 Mouse = 0.9
NBCAPEG
non-binding control adnectina +40kDa branched polyethylene glycol (PEG)
10.9 + 40 (true)
5.14
NA
500 (apparent)d
AC C
EP
TE D
(c305)
184
a
185
fibronectin; b Calculated using the GPMAW algorithm; c NA – not measured since binding to FcRn
186
not anticipated; d Apparent molecular weight; e no detectable binding.
Adnectins (e.g., NBCA) are a 10-12 kDa engineered form of the 10th repeat Type III domain of human
187 188
2.2. Study dosing and exposure parameters
8
ACCEPTED MANUSCRIPT
Sprague-Dawley male rats (IV: 66 animals, age 7-8 weeks, weight 216-283 g; inhalation: 122 animals,
190
age 7-9 weeks, weight 213-348 g) were obtained from Charles River Canada Inc. (St-Constant, QC,
191
Canada), and randomly assigned to each group in such a way that 3 animals were utilized at each time
192
point and no animal was bled more than 3-4 times depending on the time between bleeds. For IV studies,
193
a single 2 mg/kg (BTLA mAb, BSA, and IL23 scFv) or 1 mg/kg (NBCA and NBCA-PEG) dose in
194
aqueous buffer was filtered and then injected into the tail. For inhalation studies, rats were subjected to a
195
single one-hour nose-only 40 mg test protein/m3 aerosol in aqueous vehicle or vehicle alone (Table 2).
196
The test and vehicle control aerosols were generated using Pari LC Plus air-jet nebulizers.
SC
RI PT
189
M AN U
197 198
Table 2
199
Inhalation study test protein air exposure conditions. IV and Inhalation Vehicle
NBCA
PBS, pH 7.4
Measured Air Concentration (mg/m3) 42
TE D
Test Protein
IL23 scFv 20 mM Histidine and 125 mM NaCl pH 6.8
54
BSA
45
BTLA mAb
NBCAPEG
AC C
EP
20 mM citrate, 150 mM NaCl pH 6.0 buffer plus 8.3 µl dimethicone/mL 20 mM citrate, 150 mM NaCl pH 6.0 buffer
28
PBS, pH 7.4
48
Inhaled Dose (mg/kg)
MMAD+ GSDa (µm) Analytical
MMAD+ Aggregation (pre) GSD post nebulizationb (µm) HMW species Gravimetric
1.8
1.8 ± 1.9
1.9 ± 1.8
(0.7%) 7.6%
2.4
2.2 ± 2.2
(4.1%)
2.2 ± 1.9
3.9% 2.0
0.8 ± 2.0
(20.5%)
1.0 ± 1.9
24.5% 1.2
3.6 ± 1.6
(4.2%)
2.4 ± 2.0
4.5%
2.1
1.9 ± 2.3
(0.4%)
2.0 ± 2.2
0.3%
200
a
201
potential measured as high MW species by Size Exclusion Chromatography before and after 1-hour
202
nebulization.
MMAD = mass median aerodynamic diameter; GSD = geometric standard deviation;
9
b
Aggregation
ACCEPTED MANUSCRIPT
203 The inhaled dose with and without adjustment for deposited dose was calculated according to the method
205
of Alexander and colleagues [19] to determine the estimated achieved dose (Table 3). Test protein
206
aerosol concentrations and homogeneity were determined gravimetrically from an open-face
207
polyvinylidene difluoride filter and calculated from the amount of test article collected on each filter
208
sample divided by sample volume.
209
gravimetrically at initiation of exposure and every 20 minutes for the duration of the exposure. Particle
210
size distribution was performed using an 8-stage cascade impactor, which was followed by gravimetric or
211
HPLC analysis. The stability (protein degradation or aggregation) of each test protein under study
212
conditions was determined separately by collecting samples from nebulizer reservoirs before and after 60
213
minutes of nebulization and analyzing by UPLC/MS. All animal studies were done in accordance with
214
animal welfare guidelines and reviewed by an institutional animal care use committee.
RI PT
204
M AN U
SC
Actual chamber concentrations of aerosol were measured
215 Table 3
217
Estimated achieved dose calculation. [19]
TE D
216
=
= respiratory minute volume, calculatedb
EP
Where RMV (L/min)
RMV × Active Concentration x T x IF BW
= chamber concentration of active ingredient determined by chemical analysis.
AC C
Active Concentration (mg/L) T (min) IFa
= Exposure time (60 minutes) = inhalable fraction (proportion by weight of particles that are inhalable)
= mean body weight per group from the regular body weight occasion during exposure. Total body dose assuming an inhalable fraction of 100 unless stated otherwise; b 0.608 × [body weight BW (kg)
218
a
219
(kg)] 0.852 L/min. It is assumed that this parameter is unaffected by exposure to the test item.
220 221
2.3. Sample collection
10
ACCEPTED MANUSCRIPT
After inhalation and IV exposures, animals were subjected to clinical signs examinations,
223
pharmacokinetic serum collection and terminal procedures. Based on the different pharmacokinetic
224
characteristics of test proteins, the sampling time points were selected to adequately define the
225
concentration versus time curve (i.e., the absorption, distribution, and elimination phases) for each
226
protein. In addition, total duration of sampling was generally extended to at least three to five expected
227
half-lives to accurately determine the terminal elimination half-lives of test proteins. Approximately 0.3
228
mL of blood was taken from the jugular vein at each time point (Table 4) except for terminal bleed
229
samples, which were taken from the abdominal aorta. For all inhalation studies and IV studies with IL23
230
scFv and BTLA mAb, the following postmortem lung procedures and evaluations were conducted. Body
231
and lung weights were taken at necropsy. Given the likely differences in the drug disposition of test
232
proteins between lung and blood, lung PK and tissue samples were collected at either 24 hours (NBCA)
233
or 48 hours (all other proteins) post-dose to define the expected maximum concentrations. The BAL fluid
234
was collected from the left lobe on select animals by lavaging three times with 1.5 mL of ice cold PBS-
235
albumin, which was included to improve stability in certain samples. The BAL was centrifuged and the
236
supernatant was collected for immediate cell typing and quantification, lactate dehydrogenase activity
237
determination, and the remainder frozen for test protein analysis. The left lobe deplete of its BAL, was
238
frozen, homogenized at a later date for test article analysis. The remainder of the lung lobes were fixed in
239
10% neutral buffered formalin, processed with hematoxylin and eosin, and examined by microscopy.
EP
TE D
M AN U
SC
RI PT
222
AC C
240 241
Table 4
242
Pharmacokinetic sample collection times for the intravenous and inhalations studies. Treatment
IV Study Bleeding Timepoints (hours)
Inhalation Study Bleeding Timepoints (hours)
Inhalation Study BAL Timepoints (hours)
NBCA
0.03, 0.08, 0.25, 0.5, 1, 2, 4, 8
0.5, 1.0, 1.08, 1.25, 1.5, 2, 4, 5, 9
1, 25
IL23 scFv
0.03, 0.08, 0.25, 0.5, 1, 2, 4, 8
0.5, 1, 1.08, 1.25, 1.5, 2, 4, 5, 9
25, 49
11
ACCEPTED MANUSCRIPT
0.08, 0.5, 2, 4, 8, 24, 48, 72, 96
1.08, 1.5, 3, 5, 9, 25, 49, 73, 97, 121
1.08, 3, 5, 25, 49
BTLA mAb
0.5, 2, 6, 24, 48, 72, 168, 336, 504, 840, 1344
1.5, 3, 7, 25, 49, 73, 169, 337, 505, 832, 1344
1, 1.5, 3, 25, 49
NBCA-PEG
0.03, 0.5, 1, 2, 8, 24, 48, 72, 96, 168, 240, 312, 384
0.5, 1.0, 1.5, 2, 3, 9, 25, 49, 97, 145, 169
1, 25, 49, 169
RI PT
BSA
243 244
2.4. Assays for test protein, lactate dehydrogenase, immune cells, histology, FcRn binding
246
2.4.1. Test protein analysis
247
Concentrations of the test proteins were measured in serum, lung tissue, and BAL fluid using qualified
248
Meso Scale Discovery (MSD, Gaithersburg, MD) electrochemiluminescent immunosorbent assays or
249
chemiluminescent assay (for BSA). Biotinylated analyte-specific monoclonal or polyclonal antibodies
250
were immobilized on the MSD streptavidin plate as the capture reagent. Samples, calibrators and quality
251
controls were diluted in 1% BSA/PBS, 0.05% Tween 20. After incubation in the plates, unbound material
252
was washed away and the captured analytes were detected with specific detection antibodies labeled with
253
the ruthenium complex (HRP-labeled for BSA assay). Following addition of MSD read buffer,
254
luminescence intensity was measured by a MSD reader [HRP Chemiluminescent substrate was used for
255
BSA assay and luminscence was measured on Spectramax M5 (Molecular Devices)]. The concentration
256
of each protein in serum or BAL samples was calculated using a 5-parameter logistic calibration curve
257
generated from the corresponding protein calibrators. Additional assay parameters are listed in the Table
258
5.
260 261
M AN U
TE D
EP
AC C
259
SC
245
262 263 264 12
ACCEPTED MANUSCRIPT
265 266 267
RI PT
268 269 270
272
Table 5
273
Description of test protein bioanalytical parameters. Capture Absa
NBCA
Serum
BTLA mAb
NBCA-
PEG
Range (ng/mL)
a-Adnectin Rabbit pAb
a-Adnectin Rabbit pAb
4
4 - 4,000
BAL
a-Adnectin Rabbit pAb
a-Adnectin Rabbit pAb
2
4 - 4,000
Serum
a-Hu IL-23
a-VH Goat pAb
10
20 - 5,000
BAL/lung a-Hu IL-23
a-VH Goat pAb
2
10 - 1,250
Serum
a-BSA mAb
a-BSA Goat pAb
5
20 - 10,000
BAL/lung a-BSA mAb
a-BSA Goat pAb
2
31.3 - 500
a-Hu IgG mAb
a-Hu IgG Fc mAb
5
1.4 - 1,000
BAL/lung a-Hu IgG mAb
a-Hu IgG Fc mAb
2
2 - 500
Serum
EP
BSA
MRDc (fold)
AC C
IL-23 scFv
Detection Absb
M AN U
Matrix
TE D
Analyte
SC
271
Serum
a-Adnectin Rabbit pAb
a-PEG mAb
4
1 - 400
BAL
a-Adnectin Rabbit pAb
a-PEG mAb
2
0.2 - 200
274
a
All capture Abs were biotinylated; b All detection Abs were rhutenylated except BSA, which was HRP
275
conjugated; c Minimum required dilution.
276 277
2.4.2. Lactate dehydrogenase and BAL cell assay 13
ACCEPTED MANUSCRIPT
Lactate dehydrogenase activity was determined by standard redox assay. BAL immune cell types and
279
counts were measured by resuspending the BAL sample pellet into a volume of 2 mL of PBS-0.1% PBS-
280
BSA, and cells counts were performed with a hematology analyzer (Advia 120). Additionally, cytospin
281
slides were prepared, stained and a differential cell count was evaluated microscopically.
282
2.4.3. FcRn binding
283
To assess for potential differences in FcRn binding across species for BSA and BTLA mAb (compounds
284
known to bind to FcRn), Biacore surface plasmon resonance (SPR) experiments were performed on a
285
Biacore T100 instrument (Biacore/GE Healthcare). Sensor surfaces were prepared by immobilizing 30
286
µg/ml BSA or BTLA mAb in 10 mM sodium acetate pH 4.5 on a CM5 sensor chip using standard
287
ethyl(dimethylaminopropyl) carbodiimide (EDC) / N-hydroxysuccinimide chemistry, with ethanolamine
288
blocking, a running buffer of 10 mM sodium phosphate, 130 mM sodium chloride, 0.05% Tween 20
289
(PBS-T) pH 7.1. FcRn binding experiments were performed by testing various concentrations of human-
290
FcRn, cyno-FcRn, rat-FcRn or mouse-FcRn (produced in house) from 10 µM down to 0.1 µM in PBS-T
291
pH 6.0, at either 25oC or 37oC, using association time of 240 s, dissociation time of 120 s, at a flow rate of
292
30 µl/min. Surfaces were regenerated between cycles using two 10 s pulses of 50 mM Tris, 150 mM
293
NaCl, pH 8.0, at a flow rate of 30 µl/min. Binding data were fit to a 1:1 steady state model using Biacore
294
T100 evaluation software (Biacore/GE Healthcare) with KD binding affinity .
295
2.5. Pharmacokinetic Analysis
296
All pharmacokinetic parameters were calculated from a composite mean serum test protein concentration
297
versus time curve for each group only using non-compartmental methods by Phoenix WinNonlin
298
(Certara, Princeton, NJ). The mean area under the concentration versus time curve (AUC0-T or AUC∞)
299
where T was the last measurable concentration or infinity was calculated using the linear trapezoidal rule.
300
The mean maximum serum drug concentration (Cmax) and its corresponding post exposure time point
301
(Tmax) were determined from each group composite concentration data. The elimination rate constant
302
(λz) was determined by regression of the terminal log-linear portion of the curve. The apparent terminal
AC C
EP
TE D
M AN U
SC
RI PT
278
14
ACCEPTED MANUSCRIPT
303
half-life (T1/2) was calculated utilizing the equation T1/2 = ln2/λz). Inhalation bioavailability (%BA) was
304
calculated as shown below. ( ℎ (
) × Dose( ) × Dose( ℎ
) )
× 100%
RI PT
%BA =
To assess the amount of test protein unabsorbed from the bronchvaleolar space, the BAL was examined.
306
The fluid collected from each rat was normalized to the weight of left lobe of rat lung. In addition,
307
normalized drug concentrations in serum, lung, and BAL fluid were calculated by dividing the drug
308
concentrations by the respective IV dose or inhalation exposure dose levels.
309
3. Results
310
3.1. Systemic pharmacokinetic profiles following intravenous and inhalation exposures
311
The systemic mean AUC, mean Cmax, mean Tmax, and apparent terminal T1/2 of the five test proteins
312
following IV and one-hour nose-only inhalation exposures are shown in Table 6. Dose-normalized AUC
313
and Cmax after inhalation exposure were substantially lower than those after IV administration. Reliable
314
T1/2 values after inhalation exposure could not be assessed for NBCA, IL23 scFv, BSA or NBCA-PEG
315
due to insufficient time points at the terminal phase of the concentration profile or measurements below
316
the lower limit of quantitation (LLOQ; Figures 1 and 2). However, the BTLA mAb T1/2 after inhalation
317
exposure was generally comparable to that after IV administration, indicating similar systemic clearance
318
for both routes. In addition, BTLA mAb serum AUC after inhalation was greater by an order of
319
magnitude compared to the other test proteins, likely due to good serum stability and long half-life
320
presumably through FcRn binding and recycling [20]. Inhalation delivery of BSA resulted in a virtually
321
undetectable serum concentration, forcing an overestimation of %BA by assuming serum concentrations
322
equal to the LLOQ values. Overall, there was a low level of systemic BA after inhalation exposure for
323
each test protein. When compared to the respective IV study, systemic %BA was 1.4%, 0.55%, < 0.47%,
324
0.73%, and 0.05% for NBCA, IL23 scFv, BSA, BTLA mAb, and NBCA-PEG, respectively,
325
demonstrating generally similar values and thus a lack of MW dependence within this range of MWs,
326
with the exception of the pegylated NBCA-PEG.
AC C
EP
TE D
M AN U
SC
305
15
ACCEPTED MANUSCRIPT
327 328 329
RI PT
330 331 332
SC
333 334 Table 6
336
Comparison of systemic pharmacokinetic parameters after a single intravenous and 1-hour
337
nose-only inhalation exposure in rats.
M AN U
335
NBCA
IL23 scFv BSA
BTLA mAb
NBCA-PEG
10.9
30.1
67
144
500
1
2
2
2
1
6.57
4.25
410
4140
232
19.4
15.2
19.0
21.0
13.5
1.1
1.7
25
316
47
Inhaled Dose (mg/kg)
1.8
2.4
2.0
1.24
2.1
AUC0-Ta/Inhaled Dose
0.0939
0.0235
<1.94b
29.2
0.130
0.011
0.0067
<0.02
0.083
0.0048
Tmax (h)
2.0
1.5
-c
49
9.0
T1/2 (h)
5.5
<2
-
370
21
Route
MW (kDa)
Intravenous
Dose (mg/kg) a
-1
TE D
AUC0-T /Dose
-1
(µg•h•mL /mg•kg ) Cmax/Dose -1
-1
(µg•mL /mg•kg )
AC C
Inhalation
EP
T1/2 (h)
-1
-1
(µg•h•mL /mg•kg ) Cmax/Inhaled Dose -1
-1
(µg•mL /mg•kg )
Inhalation F (%)
16
ACCEPTED MANUSCRIPT
Inhaled Dose
1.4
0.55
< 0.47
0.73
0.056
5%
15.9
9.2
9.5
22.6
0.5
10%
7.9
4.6
< 4.7
11.3
0.3
RI PT
Deposited Dosed
338
a
For AUC0-T, T = infinite time or where T is the time of the last measureable concentration; b Serum BSA
339
concentrations were less than LLOQ (0.04 µg/mL) at all time points (up to 97 hours); c Value could not
340
be determined;
341
(IF) or deposited dose as described in Table 3. The IF of 5% and 10% were based on estimations from the
342
literature [31, 35].
343
3.2. Lung Clearance and Tissue Dose after IV and Inhalation Exposure
344
For the two proteins in which lung concentrations were determined after intravenous exposure, IL23 scFv
345
and BTLA mAb, both diffused from systemic circulation across the lung barrier to the lung lining fluid as
346
demonstrated by a rapid peak in lung tissue and BAL concentrations (approximately 5 minutes to 2 hours
347
post-dose) and gradual decrease thereafter (Figure 2). Interestingly, this indicated that after IV injection,
348
test proteins rapidly distribute into the well-perfused lung for both the modest MW (i.e., the 30.9 kDa
349
IL23 scFv) and large MW (i.e., the 144 kDa BTLA mAb) proteins (Table 7). Moreover, the lung
350
exposures following IV administration (i.e., combined AUC in lung tissue and BAL) of IL23 scFv and
351
BTLA mAb were less than 10% of systemic exposures (i.e., AUC in serum), demonstrating that test
352
proteins localize in the lung at relatively low levels after IV dosing (Table 7).
353
After the one-hour inhalation exposure, there was a higher overall exposure of each test protein in the
354
lung than in the systemic circulation (Figure 1, Table 7). The concentrations peaked in BAL and lung
355
tissue immediately after the inhalation period and gradually decreased thereafter except for BTLA mAb
356
(Figure 1), which remained at approximately the same concentration over the 48-hour period examined,
357
consistent with the in vivo stability of monoclonal antibodies. The unabsorbed IL23 scFV, BSA and
358
BTLA mAb concentrations in the BAL and lung tissue were roughly equivalent on a nanomolar basis.
SC
%BA calculated utilizing an inhaled dose adjusted by a 5% or 10% inhalable fraction
AC C
EP
TE D
M AN U
d
17
ACCEPTED MANUSCRIPT
However, BTLA mAb had an order of magnitude greater serum AUC thus impacting the lung
360
tissue/serum and BAL/serum ratios dramatically compared to the other test proteins. When comparing
361
the test protein concentrations normalized by dose on a molar basis, there was a trend to a decreased
362
concentration in the BAL with increased in MW (Figure 1), which could reflect increased absorption or a
363
decreased molar administered dose for the higher MW test proteins used in the study.
364
The impact of PEGylation on pharmacokinetics was examined (Table 6). Compared to the NCBA
365
protein alone, the addition of a 40 kDa PEG increased the systemic AUC/dose 35-fold and the T1/2 40-fold
366
after IV injection, while after inhalation the systemic AUC of the two proteins were equivalent and the
367
T1/2 for the pegylated form was only four-fold greater. After inhalation, notably, the BA of NBCA-PEG
368
was drastically reduced to 0.04% compared to 1.4% for NBCA. However, in the lung (as BAL), there
369
were minimal differences in PK profiles (Figure 2). The BAL kinetic profiles for NBCA and NBCA-
370
PEG mirrored the serum profile with similar apparent T1/2 (4.5 and 7.2 hours in BAL, respectively; Table
371
7).
372
It is important to note that no clear inhalation MW-relationship was observed for the lung tissue or BAL
373
exposures or the inhalation BA values for the test proteins, with the exception of the low BA for the
374
PEGylated NBCA where size (MW of 500 kDa) did appear to impact absorption (Table 6).
377 378 379 380
SC
M AN U
TE D
EP
376
AC C
375
RI PT
359
381 382 383 384 18
ACCEPTED MANUSCRIPT
385 386 387
RI PT
388 389 390
SC
391 392
394 395
M AN U
393
Table 7
397
Exposures in lung tissue, bronchioalveolar lavage (BAL) and serum after intravenous and 1-hour nose-
398
only inhalation exposure in rats.
399
A) Intravenous
0.228
BTLA mAb
74
AUC Ratio
BAL T1/2
Lung/Serum
BAL/Serum
h
0.131
4.25
0.054
0.031
1.0
110
4140
0.018
0.027
326 b
AC C
IL23 scFv
400 401
Exposure AUC0-T a/Dose µg•h•mL-1/mg•kg-1 Lung BAL Serum
EP
Proteins
TE D
396
B) Inhalation Proteins
NBCA
Exposure AUC0-T a/Inhaled Dose µg•h•mL-1/mg•kg-1 Lung BAL Serum NA
1481
AUC Ratio
Lung/ Serum NA
0.0939
19
BAL/ Serum 15800
BAL T1/2 h 4.5
ACCEPTED MANUSCRIPT
15.5
126
0.0235
661
5360
5.9
BSA
5.70
79.5
<1.94b
<2.9
<41
19.0
BTLA mAb
10
201
29.2
0.34
6.9
-c
NBCA-PEG
NA
399
0.13
NA
3070
7.2
RI PT
IL23 scFv
NA: lung tissue was not collected; a Dose normalized AUC in µg•h•mL-1/mg•kg-1 where T = infinite time
403
or where T is the time of the last measureable concentration; b Serum BSA concentrations were less than
404
LLOQ (0.04 µg/mL) at all time points (up to 97 hours); c BAL BTLA mAb concentrations did not
405
decrease between 1 and 49 hours so T1/2 could not be calculated.
M AN U
406
SC
402
407 408
3.3. Toxicological Impact of Test Protein Exposure
410
In all studies, there were no clinical signs observed that would indicate any detrimental effect of the test
411
proteins on the animals. In the inhalation studies, lung histology, BAL lactose dehydrogenase levels,
412
BAL immune cell type and counts were determined. With all proteins, minimal to no changes compared
413
to vehicle controls were observed after the one-hour inhalation exposure (data not shown). It should be
414
noted that the immunological hypersensitivity (Type I) potential, which would require chronic or repeat
415
exposure, was not assessed in this study.
416
3.4. Test protein description, physical integrity, and aerosolization of test proteins
417
The test proteins were selected to mimic typical protein therapeutics across a range of molecular weights.
418
As the binding properties at the therapeutic target may influence the pharmacokinetics, this specific target
419
binding in the rat was determined to be minimal to none for all proteins (data not shown) and thus was not
420
considered a factor for these proteins. However, the present study also assessed the FcRn binding
421
potential for both BSA and BTLA mAb, for which FcRn binding is known to play a role in protein
422
recycling and longevity in the circulation. While not all together surprising since human serum albumin
AC C
EP
TE D
409
20
ACCEPTED MANUSCRIPT
was found to have poor FcRn binding in rodents [21], BSA did not bind to the rat FcRn as compared to
424
that in the human. As expected, there was strong binding of BTLA mAb to both human and rat FcRn.
425
As particle size influences deposition and concentration in the alveolar region, the nebulizer output was
426
assessed to ensure consistency and stability of the aerosol being delivered to the rat. The test protein
427
concentration in the inhaled air-stream, mass median aerodynamic diameter (MMAD) and the aerosol
428
droplet size was consistent across all test proteins (Table 2). Aerosol particle size estimation by two
429
different methods fell within a narrow range of 0.8-3.6 µm MMAD with a low standard deviation of 1.6-
430
2.2 µm for all test proteins (Table 2). Being aqueous buffer-based formulations that aerosolize in a
431
similar fashion, they are not likely to exhibit different aerodynamic deposition profiles. Therefore,
432
deposition was excluded from the interpretation of data. Additionally, the testing from the formulation
433
reservoir at the start and end of the nebulization period indicated that the test proteins were stable (Table
434
2), with the exception of BSA where the presence of 24.5% of a high MW species suggests a strong
435
tendency to agglomerate despite the presence of an antifoaming agent in the formulation. This increased
436
percentage of MW species likely decreased the concentration of molecules available for diffusion into the
437
systemic circulation, explaining the lack of detectable BSA in the serum after inhalation exposure.
438
TE D
M AN U
SC
RI PT
423
4. Discussion
440
4.1. Key findings
441
To more accurately determine the OEL and control workplace exposures of protein therapeutics, the fate
442
and disposition of proteins of different molecular weights and complexity were explored after inhalation
443
and IV exposure. The strategy was to determine the %BA of aerosolized proteins in a rat inhalation
444
model simulating nuisance dust air concentrations (e.g., PNOS – particles not otherwise specified [22]),
445
as a “worst case scenario” and utilizing an exposure length representing the time for typical open
446
processing in biologics manufacturing.
447
examining test proteins covering a MW range from 10 – 150 kDa, suggested a low systemic BA of
448
approximately 1% or less is reached after inhalation exposure. Second, from examination of lung and
AC C
EP
439
We identified three key findings.
21
First, the results from
ACCEPTED MANUSCRIPT
BAL, the test proteins were present at similar concentrations in the lung over the 48-hour observation
450
period, when considered on a molar basis. While negligible adverse impact to the integrity of the lung
451
was observed, there could be the potential for accumulation in the lung and low sustained systemic
452
release if there are repeated exposures. This could also allow residual protein to interact with therapeutic
453
target lung receptors, which was reported with the lung accumulation of cetuximab (152 kDa, IgG1
454
antibody for the treatment of cancer) after pulmonary delivery to mice [9, 23]. Lastly, following the fate
455
of the test proteins in the lung after IV exposure, there was clear evidence of the test proteins in the lung,
456
indicating that a low percentage, <10% of systemic AUC, of these higher MW proteins can pass from
457
circulation into BAL. Understanding that there is reasonable distribution of a test protein to the lung from
458
an IV exposure can provide an indication of potential for direct lung effects after an inhalation exposure.
459
4.2. Comparison to literature including mechanism of transfer
460
There have been a number of investigations on systemic BA of proteins after lung exposure; however
461
there is a lack of a solid consensus on a %BA value for proteins and understood absorption dependencies.
462
Several authors have provided an analysis of the literature focusing on MW with BA as a dependent
463
variable and have indicated an inverse relationship [11, 24-26]. More recently, Pfister and colleagues [6]
464
reinforced the MW dependence and provided further clarity by concluding that proteins greater than 40
465
kDa would not likely exhibit a systemic BA greater than 5% following lung exposure. The results of the
466
present study show that indeed proteins of >40 kDa and even down to 10.9 kDa have a BA lower than 5%
467
after true inhalation exposure. However, a clear difference in BA for test proteins in the MW range of 10
468
to 150 kDa was not demonstrated. The lack of a MW dependency can be explained by looking at the
469
mechanism of transfer. It is believable that as proteins increase in size, at some point, they would exceed
470
the pore size to transverse the alveolar junctions. The translocation being based on restricted, size-
471
dependent passive diffusion of molecules through aqueous pores that range in size from 5 to 20 nm [27,
472
28] wherein larger molecules experience greater hindrance entering and navigating across the alveolar
473
epithelial barrier. With the test proteins examined under the same experimental conditions, there were
474
low but similar test protein concentrations on a molar basis in lung tissue, BAL and serum (excluding the
AC C
EP
TE D
M AN U
SC
RI PT
449
22
ACCEPTED MANUSCRIPT
pegylated protein) (Table 7), thus supporting the concept that passive paracellular or transcellular
476
transport from the airways to vasculature is generally minimal. Looking at BTLA mAb absorption, the
477
data presented here do not differentiate whether absorption occurred only by the expected receptor
478
mediated (i.e., FcRn) active transport, passive diffusion, or a combination of both. Regardless, the low
479
systemic absorption of a full antibody with an active FcRn region following inhalation exposure was
480
similar to the presumed passive transport for the lower molecular weight test proteins. Guilleminault and
481
colleagues showed that passive diffusion can occur after intratracheal exposure of a Fc containing
482
antibody in a FcRn knockout mouse; while there was low systemic BA (7.9%), the overall exposure was
483
8% of that in the wildtype mouse under the same conditions [9]. While a definitive MW cut off for
484
epithelial membrane transfer was not examined, it can clearly be seen that the absorption drops off
485
dramatically with NSCA-PEG (apparent size of approximately 500 kDa), which demonstrated a BA of
486
0.05% as compared to the other proteins examined in this study. Other mechanisms of entry such as
487
through the lymphatic system may be in play in this case. Overall, there appears to be a plateau in
488
systemic BA after inhalation exposure for proteins in the MW range of 10 to 150 kDa.
489
The literature has sparse BA examples for proteins after lung exposure and the BA values are larger than
490
those observed in the present study. Proteins in the 9.4 – 22 kDa range have been reported to have much
491
higher BA at 10 – 56%; and as the MW increased to between 37 – 150 kDa, the BA decreased, and
492
appeared to fall into the < 5% range [11, 29, 30], supporting Pfister and colleagues’ conclusion that there
493
is a MW dependency. This elevated BA can be explained because these studies were conducted utilizing
494
intratracheal administration. We suspect the primary reason for the discrepancy between the approximate
495
1% BA of the present study and these reports is that intratracheal instillation represents a total deposited
496
dose; whereas here, the %BA is provided as an inhaled dose. A deposited dose was also calculated
497
(Table 6) and applied to an inhaled dose but multiple methods are available and depending on particle
498
size, the deposited dose could vary from <5% to 10% deposited in the rat pulmonary region [31-33],
499
impacting the BA result considerably (Table 6; e.g., NCBA inhaled %BA = 1.4% compared to NCBA
500
deposited %BA of 8% -16% assuming a 5% or 10% deposited dose, respectively). Thus the methodology
AC C
EP
TE D
M AN U
SC
RI PT
475
23
ACCEPTED MANUSCRIPT
for selecting the deposited dose would lead to varying BA increasing the uncertainty. It should be noted
502
that the %BA from the calculated deposited dose from the present study is greater than those %BA
503
published in the literature from similar sized proteins, suggesting that the calculated intratracheal %BA
504
may underestimate the true inhaled %BA. Overall, this comparison underscores the difference between
505
utilizing passive inhalation versus deposited intratracheal administration for understanding lung exposure,
506
indicating most results from the literature cannot be directly compared to the current study findings.
507
A similar sized protein to IL23 scFv of this work, a 29 kDa protein, dornase alpha (recombinant human
508
deoxyribonuclease I for the treatment of mucous accumulation in cystic fibrosis patients) was
509
administered to humans by nebulizer and pharmacological action in the lung was demonstrated. While
510
not specifically providing a %BA, there was no measurable change in background systemic DNAse
511
concentrations after three 10 mg doses for 6 consecutive days [34], behaving pharmacokinetically similar
512
to the low BA and short half-life observed with our 30.9 kDa protein, IL23 scFv.
513
Next, in looking at the BSA results, surprisingly, it was not measurable in the serum after inhalation
514
exposure. This is in contrast to Folkesson and colleagues’ [35] report of a BA of 6.4% (note: lung
515
deposition was incorporated in their %BA value), based on an one-hour nose-only inhalation exposure of
516
33 mg BSA/m3. This difference can be explained by first, the low serum concentration in the present
517
study due to a substantial percentage of a large molecular weight species reducing systemic absorption;
518
and, secondly, the lack of bovine albumin binding to the rat FcRn; both would minimize passive or active
519
transport translocation into the circulation.
520
Scientists have shown that Fc fusion proteins have enhanced absorption as compared to proteins of
521
equivalent size without an Fc region. The BA after intratracheal aerosol instillation of erythropoietin-Fc
522
fusion protein (112 kDa protein, functions to stimulate erythrocyte production) was 35%, which is much
523
higher than observed with any test protein in the current study. It is recognized that the %BA value of
524
erythropoietin-Fc fusion protein included an adjustment for 12.8% deposition [15]. With another protein,
525
an IgG antibody omalizumab (149 kDa; anti-IgE antibody for the treatment of asthma), Pfister and
526
colleagues [6] calculated the systemic BA from a published human nebulizer study [36]. The BA was
AC C
EP
TE D
M AN U
SC
RI PT
501
24
ACCEPTED MANUSCRIPT
527
low at 0.2 - 0.6% and in the same range as our BTLA mAb, even though those studies were designed to
528
optimize inhalation delivery as an inhaled product.
529
cetuximab (152 kDa, IgG1 chimeric antibody for the treatment of cancer) to mouse and non-human
530
primate resulted in what was described as low systemic exposure (i.e., 3.9% or 0.3%, respectively). In
531
addition, a similar persistent presence of cetuximab in the lung [9, 23] was observed in agreement with
532
the BTLA mAb lung clearance.
533
Finally, the addition of a large PEG group (40kDa) to the test protein NBCA drastically reduced the BA.
534
This observation is consistent with McLeod and colleagues’ [17] who reported that systemic BA after
535
intratracheal instillation of a 60 kDa PEGylated 19 kDa interferon protein was extremely low (< 0.4%).
536
Overall, the present study increases the body of evidence to support that the lung epithelium serves as an
537
effective barrier for minimizing the transport of proteins.
538
4.3. Evaluation of %BA value for human risk assessment
539
The present studies were conducted for human risk assessment, specifically to identify possible default
540
value(s) for the route-to-route modifying factor, α in the OEL calculation for proteins. The study design
541
was optimized with human exposure in mind. The target particle size of 2.0 µm maximized deep lung
542
deposition as well as an air concentration equivalent to the highest allowable level for particles of low
543
hazard concern (Particles Not Otherwise Specified –PNOS) according to American Conference of
544
Governmental Industrial Hygienists [22, 33, 37]. This 40 mg/m3 test protein aerosol concentration
545
provided to the rat included the standard adjustment of 4x to obtain a human equivalent deposited dose to
546
the pulmonary region [38, 39]. With a maximized concentration in the alveolar space (i.e., BAL),
547
according to Fick’s first law of diffusion, optimal test item flow into the circulation would be expected,
548
assuming similar mechanism for transfer across membranes between humans and rats. In addition, non-
549
binding proteins, with the exception of the FcRn binding of BTLA mAb, were selected to minimize
550
pharmacological dependent impact on the measured pharmacokinetics. Another key factor in accepting
551
this study’s %BA value is that this was an inhalation study. General practice in risk assessment for
552
inhalation studies is to utilize a minimal to no conversion factor for interspecies for rat to human
AC C
EP
TE D
M AN U
SC
RI PT
Similarly, intratracheal aerosolized delivery of
25
ACCEPTED MANUSCRIPT
adjustment for local lung effects [38].
Likewise, the BA determined from the inhaled dose is
554
representative of occupational exposure and can be equated to the critical effect dose (Point of Departure)
555
utilized for the OEL determination where the predicted air concentration and the amount the individual
556
inhales over time comprises a human inhaled dose. While there is possibly some influence of MW at the
557
lower MW range of the test proteins examined and the differing mechanisms of movement from the lung
558
into the systemic circulation, the variability between the resulting BA values do not represent a
559
biologically meaningful difference.
560
supporting literature, we postulate that a default α of 1% to account for inhalation BA is reasonable for
561
similar protein therapeutics in the MW range of 10 – 150 kDa.
562
4.4. Impact to OEL calculation
563
Changing the absorption factor (α) in the OEL calculation can provide a dramatic difference in outcome.
564
For example, assuming all other factors and adjustments were the same, the application of an α of 100%
565
(default for no inhalation data), 5% [6] or 1% (present study), a protein therapeutic OEL would differ
566
from 1 µg/m3 to 20 µg/m3 to 100 µg/m3, respectively. Utilizing a protein therapeutic default approach
567
rather than the worst-case scenario of 100% may have far-reaching advantages. The National Institute of
568
Occupational Safety and Health (NIOSH) has described one characteristic for placing a drug on the
569
Hazardous Drug Alert List as having an OEL at ≤ 10 µg/m3. The decreased intrinsic hazard due to the
570
limited ability of proteins to be systemically bioavailable via inhalation as well as negligible through
571
dermal and oral routes should be considered when describing drugs as hazardous in this context; thus
572
minimizing application of excessive warnings to molecules of lesser risk and focusing efforts on drugs of
573
serious concern. Additionally, an increased OEL value, while ensuring protection of human health, may
574
also eliminate potentially unnecessary exposure controls, the ability of a facility to meet the OEL and
575
consequently, greater flexibility in manufacturing decisions.
576
4.5. Caveats to use of 1%
577
The study here chose model proteins with minimal interaction with the rat physiology with the purpose of
578
confirming the systemic BA of proteins after inhalation exposure. While the data presented here is based
RI PT
553
AC C
EP
TE D
M AN U
SC
In light of these data presented here in conjunction with the
26
ACCEPTED MANUSCRIPT
on one representative protein of each size or type, it was assumed that proteins of similar size whether
580
native, modified or conjugated would have similar fate and BA in a normal human lung environment
581
experiencing mucociliary clearance, protease inactivation, physical entrapping and macrophage engulfing
582
of inhaled material, all of which may influence the extent of absorption. The systemic BA assumptions
583
presented here may not be applicable to all types of biologic therapeutics and would not be relevant when
584
conducting human risk assessment on proteins that have a MW of less than 10 kDa, have direct effects on
585
the lung (e.g. receptor-mediated pharmacology, respiratory sensitization - Type 1 hypersensitivity),
586
atypical protein chemistry (e.g., polycationic chain), or other biological materials (e.g., oligonucleutides,
587
RNA, DNA, viruses, etc). Additional studies may be needed to better understand specific circumstances.
588
Nonetheless, this study outlines a default BA criteria and recommendation in pursuing OEL estimation
589
for protein therapeutics, especially for compounds in pharmaceutical product development where a
590
limited body of knowledge is available.
591
5. CONCLUSION
592
Although there are several mechanisms to hinder proteins from entering the systemic circulation via
593
inhalation, some fraction of the amount of a protein that enters the lung is expected to become
594
bioavailable. Many of the experiments reported in the literature were conducted using conditions to
595
optimize deposition and enhance absorption. The method of delivery, inhalation device used, the type of
596
formulation (nebulized liquid or passively inhaled dry powder), intersubject variability, or the
597
pathophysiological state of the lung etc., all contribute to the BA of a protein. Since information on these
598
factors or their potential impact on inhalation BA in an occupational setting is limited, this study set out to
599
better understand fate and distribution of protein therapeutics ranging in MW from 10 to 150 kDa after
600
inhalation exposure.
601
approximately 1% or less. These pharmacokinetic properties suggest that compared to small molecule
602
drugs, there is an intrinsic decrease in hazard for proteins and the application of a default pharmacokinetic
603
factor to the OEL calculation should be considered for typical proteins. This estimate is expected to be
604
conservative and protective for the worker population. Furthermore, although in this study there was
AC C
EP
TE D
M AN U
SC
RI PT
579
Overall, systemic bioavailability after inhalation exposure was low, at
27
ACCEPTED MANUSCRIPT
605
negligible to minimal lung effects from the test protein based on local evaluation, direct effects on the
606
lung should be considered when assessing the hazard potential for specific therapeutic proteins that have
607
lung targets.
RI PT
608 Conflicts of interest
610
The authors declare that they are employees of Bristol-Myers Squibb and the study was sponsored by
611
Bristol-Myers Squibb.
SC
609
612 Acknowledgements
614
Funding for this project was provided by Bristol-Myers Squibb. We would like to thank the scientists at
615
Bristol-Myers Squibb who supported this study: Guodong Chen, Sybille Herzer, Noah Ditto, Robert Li,
616
Mark Maurer, Thomas McDonagh, Jingjie Mo, Siegfried Rieble, Mark Rixon, Lumelle Schneeweis,
617
Lakshmi Sivaraman, Sally Thompson-Iritani, Caren Villano, Jack Valentine, Chunlei Wang, Hui Wei,
618
Hua Yao, Yan Yao, Chuan Zheng, Mian Gao, and Lin Cheng. In addition, the individuals responsible for
619
the conduct of the studies: Marilyne Boyer Johanne Laporte, Marie-Eve Rodrigue, Bassem Attalla, and
620
Sandra Gagnon.
EP
621
TE D
M AN U
613
Figure legends
623
Figure 1. Comparison of dose-normalized mean concentrations (nM/mg•kg-1) in lung tissue (closed
624
triangle) and bronchioalveolar lavage (open square) with those in serum (closed circle) after a 1-hour nose
625
only inhalation exposures in rats with NBCA (A), IL23 scFv (B), BSA (C), BTLA mAb (D), and NBCA-
626
PEG (E). Serum BSA concentrations were < LLOQ at all time points in Panel C. Lung tissue NBCA and
627
NBCA-PEG concentrations were not assessed in Panels A and E. Units are presented in molar quantities
628
normalized by dose in order to account for differences in molecular weight and dose.
629
Figure 2. Comparison of dose-normalized mean concentrations (nM/mg•kg-1) in lung tissue (closed
630
triangle) and bronchioalveolar lavage (open square) with those in serum (closed circle) after
AC C
622
28
ACCEPTED MANUSCRIPT
631
administration of a single IV dose in rats with IL23 scFv (A) and BTLA mAb (B). Units are presented in
632
molar quantities normalized by dose in order to account for differences in molecular weight and dose.
633
RI PT
634 635 636
SC
637 638
640 641 642 643 Appendix A. Supplemental Data
TE D
644
M AN U
639
645 Table A.1
647
Comparison of systemic pharmacokinetic parameters (in molar units) after a single intravenous and 1-
648
hour nose-only inhalation exposure in rats.
EP
646
AC C
649
NBCA IL23 scFv BSA
BTLA mAb
NBCA-PEG
Route
MW (kDa)
10.9
30.1
67
144
500
Intravenous
Dose (mg/kg)
1
2
2
2
1
603
141
6120
28800
464
1780
505
284
146
27.0
AUC0-Ta/Dose -1
(nM•h/mg•kg ) Cmax/Dose -1
(nM/mg•kg )
29
ACCEPTED MANUSCRIPT
1.1
1.7
25
316
47
Exposure Dose (mg/kg)
1.8
2.4
2.0
1.24
2.1
AUC0-Ta/Exposure Dose
8.61
0.78
<28.96
1.01
0.22
<0.30
Tmax (h)
2.0
1.5
-b
T1/2 (h)
5.5
short
-
F (%)
1.4
0.55
(nM•h/mg•kg-1) Cmax/Exposure Dose
a
651
not be determined.
< 0.47
M AN U
650
0.58
0.26
0.010
49
9.0
370
21
0.73
0.056
SC
(nM/mg•kg-1)
202
RI PT
Inhalation
T1/2 (h)
For AUC0-T, T = infinite time or where T is the time of the last measureable concentration; b Value could
652
AC C
EP
TE D
653
30
ACCEPTED MANUSCRIPT
654
References
655
1.
Serebrov, M., Biologics' share of medicine chest grows, and so do the pricing concerns. 2016.
656
2.
Administration, F.a.D. What Are "Biologics" Questions and Answers. 2015; Available from: http://www.fda.gov/AboutFDA/CentersOffices/OfficeofMedicalProductsandTobacco/CBER/ucm
658
133077.htm.
659
3.
RI PT
657
Bercu, J.P., et al., Point of departure (PoD) selection for the derivation of acceptable daily
exposures (ADEs) for active pharmaceutical ingredients (APIs). Regul Toxicol Pharmacol, 2016.
661
79 Suppl 1: p. S48-56. 4.
exposure limits for pharmaceuticals. Regul Toxicol Pharmacol, 2016. 79 Suppl 1: p. S67-78.
663 664
Reichard, J.F., et al., Toxicokinetic and toxicodynamic considerations when deriving health-based
M AN U
662
SC
660
5.
Sussman, R.G., et al., A harmonization effort for acceptable daily exposure derivation -
665
Considerations for application of adjustment factors. Regul Toxicol Pharmacol, 2016. 79 Suppl
666
1: p. S57-66.
Occup Hyg, 2014. 58(7): p. 899-911.
668 7.
Nat Rev Drug Discov, 2007. 6(1): p. 67-74.
670 671
8.
9.
10.
678
Depreter, F., G. Pilcer, and K. Amighi, Inhaled proteins: challenges and perspectives. Int J
Pharm, 2013. 447(1-2): p. 251-80.
676 677
Guilleminault, L., et al., Fate of inhaled monoclonal antibodies after the deposition of aerosolized
particles in the respiratory system. J Control Release, 2014. 196: p. 344-54.
674 675
Patton, J.S., J.G. Bukar, and M.A. Eldon, Clinical pharmacokinetics and pharmacodynamics of inhaled insulin. Clin Pharmacokinet, 2004. 43(12): p. 781-801.
672 673
Patton, J.S. and P.R. Byron, Inhaling medicines: delivering drugs to the body through the lungs.
EP
669
Pfister, T., et al., Bioavailability of therapeutic proteins by inhalation--worker safety aspects. Ann
TE D
6.
AC C
667
11.
Patton, J.S., C.S. Fishburn, and J.G. Weers, The lungs as a portal of entry for systemic drug delivery. Proc Am Thorac Soc, 2004. 1(4): p. 338-44.
31
ACCEPTED MANUSCRIPT
679
12.
NIOSH, National Institute for Occupational Safety and Health. (2016) NIOSH Pocket Guide to
680
Chemical Hazards - Nickel metal and other compounds. Center for Disease Control. Last
681
reviewed 11 April 2016. [online]. Available at: https://www.cdc.gov/niosh/npg/npgd0445.html. 13.
receptor, FcRn. J Immunol, 2004. 172(4): p. 2021-9.
683 684
Ober, R.J., et al., Visualizing the site and dynamics of IgG salvage by the MHC class I-related
14.
RI PT
682
Spiekermann, G.M., et al., Receptor-mediated immunoglobulin G transport across mucosal barriers in adult life: functional expression of FcRn in the mammalian lung. J Exp Med, 2002.
686
196(3): p. 303-10. 15.
Bitonti, A.J., et al., Pulmonary delivery of an erythropoietin Fc fusion protein in non-human
M AN U
687
SC
685
688
primates through an immunoglobulin transport pathway. Proc Natl Acad Sci U S A, 2004.
689
101(26): p. 9763-8. 16.
receptor-mediated transcytosis. Hum Reprod, 2005. 20(7): p. 1805-13. 17.
following pulmonary administration. J Pharm Sci, 2015. 104(4): p. 1421-30.
693 18.
calcitonin: optimization by PEG size selection. J Control Release, 2008. 125(1): p. 68-75.
695 696
19.
inhalation toxicology studies with pharmaceuticals. Inhal Toxicol, 2008. 20(13): p. 1179-89.
698 20.
Roopenian, D.C. and S. Akilesh, FcRn: the neonatal Fc receptor comes of age. Nat Rev
Immunol, 2007. 7(9): p. 715-25.
700 701
Alexander, D.J., et al., Association of Inhalation Toxicologists (AIT) working party recommendation for standard delivered dose calculation and expression in non-clinical aerosol
697
699
Youn, Y.S., et al., Improved intrapulmonary delivery of site-specific PEGylated salmon
EP
694
McLeod, V.M., et al., Optimal PEGylation can improve the exposure of interferon in the lungs
TE D
691 692
Low, S.C., et al., Oral and pulmonary delivery of FSH-Fc fusion proteins via neonatal Fc
AC C
690
21.
Andersen, J.T., et al., Single-chain variable fragment albumin fusions bind the neonatal Fc
702
receptor (FcRn) in a species-dependent manner: implications for in vivo half-life evaluation of
703
albumin fusion therapeutics. J Biol Chem, 2013. 288(33): p. 24277-85.
32
ACCEPTED MANUSCRIPT
704
22.
ACGIH, American Conference of Governmental Industrial Hygienists) (2015). TLVs and BEIs
705
Based on the Documentation of the Threshold Limit Values for Chemical Substances and
706
Physical Agents & Biological Exposure Indices. .
tumors. Pharm Res, 2011. 28(9): p. 2147-56.
708 24.
Rev Respir Dis, 1983. 127(5 Pt 2): p. S59-65.
710 711
Effros, R.M. and G.R. Mason, Measurements of pulmonary epithelial permeability in vivo. Am
25.
SC
709
Maillet, A., et al., The airways, a novel route for delivering monoclonal antibodies to treat lung
RI PT
23.
Hastings, R.H., et al., Clearance of different-sized proteins from the alveolar space in humans and rabbits. J Appl Physiol (1985), 1992. 73(4): p. 1310-6.
712
M AN U
707
713
26.
Wall, D.A., Pulmonary Absorption of Peptides and Proteins. Drug Delivery, 1995. 2(1): p. 1-20.
714
27.
Matsukawa, Y., et al., Size-dependent dextran transport across rat alveolar epithelial cell monolayers. J Pharm Sci, 1997. 86(3): p. 305-9.
715 28.
29.
Reviews, 1996. 19(1): p. 3-36. 31.
Green, J.D., Pharmaco-toxicological expert report Pulmozyme rhDNase Genentech, Inc. Hum Exp Toxicol, 1994. 13 Suppl 1: p. S1-42.
723 724
32.
725
33.
ARA Inc, Multiple-Path Particle Dosimetry Model (MPPD) version 2.11. Arlington, Va. 2015. Raabe, O.G., et al., Regional deposition of inhaled monodisperse coarse and fine aerosol
particles in small laboratory animals. Ann Occup Hyg, 1988. 32(Supplement 1): p. 53-63.
726 727
Patton J.S., Mechanisms of macromolecule absorption by the lung. Advanced Drug Delivery
EP
30.
721 722
Schanker LS, B.J., Absorption of heparin and cyanocobalamin from the rat lung. Proc Soc Exp Biol Med. , 1976. 152(3): p. 377-80.
719 720
TE D
hetastarch absorption in isolated rat lungs. J Appl Physiol (1985), 2001. 91(4): p. 1730-40.
717 718
Conhaim, R.L., et al., Transport properties of alveolar epithelium measured by molecular
AC C
716
34.
Genentech, PULMOZYME- dornase alfa solution. Genentech, Inc. [FDA] Food and Drug
728
Administration Label information. Available at: https://dailymed.nlm.nih.gov/dailymed/index.cfm.
729
Revised December, 2014. 2014. 33
ACCEPTED MANUSCRIPT
730
35.
Folkesson HG, W.B., Dahlbäck M, Lundin S, Karlsson BW., Passage of aerosolized BSA and the
731
nona-peptide dDAVP via the respiratory tract in young and adult rats. Exp Lung Res, 1992.
732
18(5): p. 595-614.
asthmatic subjects. Am J Respir Crit Care Med, 1999. 160(3): p. 1023-7.
734 735
37.
Schlesinger, R.B., Comparative deposition of inhaled aerosols in experimental animals and humans: a review. J Toxicol Environ Health, 1985. 15(2): p. 197-214.
736 737
Fahy, J.V., et al., Effect of aerosolized anti-IgE (E25) on airway responses to inhaled allergen in
RI PT
36.
38.
SC
733
ECETOC, Derivation of Assessment Factors for Human Health Risk Assessment. Techinical Report No. 86. European Centre for Ecotoxicology and Toxicology of Chemicals. Brussels,
739
Belgium. ISSN-0773-6347-86. Available online: http://www.ecetoc.org/wp-
740
content/uploads/2014/08/ECETOC-TR-086.pdf. 2003.
741
39.
M AN U
738
EPA, Methods for Derivation of Inhalation Reference Concentrations and Application of Inhalation Dosimetry. U.S. Environmental Protection Agency. Research Triangle Park, North
743
Carolina. EPA/600/8-90/066F. Available online:
744
https://www.epa.gov/sites/production/files/2014-11/documents/rfc_methodology.pdf. 1994.
747 748 749 750 751
EP
746
AC C
745
TE D
742
752 753 754 755 34
ACCEPTED MANUSCRIPT
(A) NBCA
M AN U
SC
RI PT
(B) IL23 scFv
(C) BSA
AC C
(E) NBCA-PEG
EP
TE D
(D) BTLA mAb
1
Figure 1 1
ACCEPTED MANUSCRIPT
(B) BTLA mAb
EP
TE D
Figure 2
AC C
2
M AN U
SC
RI PT
(A) IL23 scFv
2
ACCEPTED MANUSCRIPT
Highlights •
For typical large proteins (10-150 kDa), systemic bioavailability in the rat following inhalation is low, at ~≤1%. Addition of a PEG moiety on a protein can considerably decrease systemic bioavailability.
•
Direct effects on the lung should be considered when assessing the hazard potential for proteins
RI PT
•
that have lung targets.
SC
The application of a default PK factor adjusting for 1% bioavailability is warranted when
EP
TE D
M AN U
calculating OELs.
AC C
•