Journal Pre-proof Nonclinical safety of tildrakizumab, a humanized anti–IL-23p19 monoclonal antibody, in nonhuman primates Michael Santostefano, Danuta Herzyk, Diana Montgomery, Jayanthi Wolf PII:
S0273-2300(19)30240-5
DOI:
https://doi.org/10.1016/j.yrtph.2019.104476
Reference:
YRTPH 104476
To appear in:
Regulatory Toxicology and Pharmacology
Received Date: 1 May 2019 Revised Date:
18 July 2019
Accepted Date: 13 September 2019
Please cite this article as: Santostefano, M., Herzyk, D., Montgomery, D., Wolf, J., Nonclinical safety of tildrakizumab, a humanized anti–IL-23p19 monoclonal antibody, in nonhuman primates, Regulatory Toxicology and Pharmacology (2019), doi: https://doi.org/10.1016/j.yrtph.2019.104476. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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. © 2019 Published by Elsevier Inc.
1
Nonclinical safety of tildrakizumab, a humanized anti–IL-23p19 monoclonal
2
antibody, in nonhuman primates
3
Michael Santostefanoa, Danuta Herzykb, Diana Montgomeryc, Jayanthi Wolfd
4
a
5
Pasteur, Boston, MA, 02115-5727, United States;
[email protected]
6
b
7
Pike, West Point, PA, 19486, United States;
[email protected]
8
c
9
Pike, West Point, PA, 19486, United States;
[email protected]
Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 33 Avenue Louis
Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 770 Sumneytown
Pharmacokinetics, Predictive and Clinical Immunogenicity, Merck & Co., Inc., 770 Sumneytown
10
d
11
2505, United States;
[email protected]
Global Regulatory Affairs, Merck & Co., Inc., 351 N. Sumneytown Pike, North Wales, PA 19454-
12 13
Corresponding author:
14
Michael Santostefano
15
Tel: (617) 992-3030
16
Fax: (617) 992-2487
17 18
Abstract word count: 196
19
Text word count: 4475
20
Reference word count: 1434
21
1
22
Highlights
23
•
24 25
psoriasis in the US, EU, and Australia •
26 27
•
32
Tildrakizumab was well tolerated, with no toxicological findings in repeat-dose toxicity or embryofetal development studies
•
30 31
Safety of subcutaneously administered tildrakizumab was characterized using a pharmacologically relevant cynomolgus monkey
28 29
The anti–IL-23p19 mAb tildrakizumab is approved for moderate-to-severe plaque
No neonatal deaths were noted following in utero exposure at 7x the recommended human dose
•
The results of this comprehensive nonclinical safety program support the safe use of tildrakizumab
33
2
34 35
ABSTRACT Tildrakizumab (also known as MK-3222), is a high-affinity, humanized, immunoglobin
36
G1κ monoclonal antibody targeting the p19 subunit of interleukin-23 recently approved for
37
the treatment of moderate to severe plaque psoriasis in the US, Europe, and Australia. The
38
safety profile of tildrakizumab was characterized in nonclinical studies using a
39
pharmacologically relevant cynomolgus monkey model. In repeat-dose toxicity studies,
40
cynomolgus monkeys were chronically treated with subcutaneous (SC) injections of
41
100 mg/kg of tildrakizumab every 2 weeks up to 9 months. Tildrakizumab was well tolerated,
42
with no toxicological findings (including assessment of reproductive organs; hormonal effects;
43
and cardiovascular, respiratory, and central nervous system function) at systemic exposures
44
approximately 90 times higher than the recommended human dose of 100 mg. An
45
embryofetal developmental study conducted in pregnant monkeys revealed no treatment-
46
related effects to the developing fetus following SC administration of tildrakizumab
47
100 mg/kg. In a pre- and postnatal development study, 2 neonatal deaths due to potential
48
viral infection at 100 mg/kg were considered of uncertain relationship to the treatment based
49
on a lack of historical data on the occurrence of viral infection in neonate cynomolgus
50
monkeys. The results of this comprehensive nonclinical safety program support the safe use
51
of tildrakizumab.
52 53
KEYWORDS
54
Monoclonal antibody; interleukin-23; psoriasis; pre- and postnatal development; animal model
55 56
3
57 58
1. INTRODUCTION Psoriasis is a chronic, noncommunicable, inflammatory skin disorder with a prevalence
59
of 0.09% to 11.4% worldwide (World Health Organization). Plaque psoriasis (psoriasis
60
vulgaris)—the most common form—affects up to 97% of all patients (Kubota et al., 2015) and
61
is characterized by recurrent episodes of inflammatory, sharply demarcated, erythematous,
62
scaly plaques of variable size and confluence (World Health Organization). The primary goal of
63
psoriasis therapies is to maintain control of the lesions by clearing psoriatic plaques, which is
64
often defined in clinical trials by a 75% improvement from baseline based on the Psoriasis Area
65
and Severity Index (Fredriksson and Pettersson, 1978; Mease, 2011).
66
Currently approved biological treatments for psoriasis include tumor necrosis factor -α
67
antagonists (Enbrel® [etanercept]. Full prescribing information, 2017; Humira® [adalimumab]
68
full prescribing information, 2018; Remicade® [infliximab] full prescribing information, 2018); an
69
interleukin (IL)-12/23p40 antagonist (Stelara® [ustekinumab] full prescribing information,
70
2018); IL-17 antagonists (Cosentyx® [secukinumab] full prescribing information, 2018; Siliq™
71
[broadalumab] full prescribing information, 2017; Taltz® [ixekizumab] full prescribing
72
information, 2018); and IL-23p19 antagonists (Ilumya™ [tildrakizumab] full prescribing
73
information, 2018; Tremfya® [guselkumab] full prescribing information, 2017).
74
IL-23 is a naturally occurring heterodimeric cytokine that shares the IL-12/23p40 subunit
75
with IL-12. The IL-12/23p40 subunit couples with either the IL-23p19 or IL-12p35 subunits to
76
form IL-23 and IL-12, respectively (Teng et al., 2015). Until the discovery of IL-23, the IL-
77
12/23p40 subunit was believed to be unique to IL-12, stimulating efforts to understand the
78
relative roles of these 2 cytokines in immune modulation (Teng et al., 2015). IL-23 is a
79
proinflammatory cytokine that plays a role in the pathogenesis of immune diseases including
80
psoriasis, ankylosing spondylitis, and psoriatic arthritis (Teng et al., 2015); a specific
4
81
hypomorphic polymorphism in the gene coding for IL-23 receptor (rs11209026) was identified
82
by genome-wide association studies as protective against these diseases (Abdollahi et al.,
83
2016). IL-23 is overexpressed in psoriasis tissue biopsies (Boutet et al., 2018; Tonel et al.,
84
2010). Sequence variants in the genes for IL-23R and its ligand confer protection against
85
psoriasis (Abdollahi et al., 2016; Capon et al., 2007). The R381Q protective allele attenuates IL-
86
23R signaling in healthy donors and psoriasis patients in in vitro functional studies (Di Meglio et
87
al., 2011).
88
Treatment of inflammatory diseases with immunosuppressive agents carries a potential
89
unintended consequence of impaired host-defense and/or tumor surveillance (Bongartz et al.,
90
2006; Davies et al., 2013; Langley et al., 2013; Teng et al., 2015). However, selective inhibition
91
of the IL-23p19 pathway is associated with decreased infection liability and decreased risk of
92
tumor incidence or progression compared to broader IL-12/23p40 neutralization (Belladonna et
93
al., 2006; Chackerian et al., 2006; Held et al., 2008; Kapil et al., 2009; Khader et al., 2005;
94
Kortylewski et al., 2009; Langowski et al., 2006; Rudner et al., 2007; Schulz et al., 2008; Teng
95
et al., 2010; Teng et al., 2012; Teng et al., 2011; von Scheidt et al., 2014; Wu et al., 2007; Wu
96
et al., 2009; Zelante et al., 2007). Animal disease model studies point to a critical role of the IL-
97
23 pathway in the pathogenesis of psoriasis, suggesting that IL-23 is an important target for
98
therapeutic intervention in the treatment of inflammatory diseases (Cua et al., 2003; Langrish
99
et al., 2005; Teng et al., 2015; Teng et al., 2011; Tonel et al., 2010) with potential for reduced
100 101
risk for infections and carcinogenesis compared to broader IL-12/23p40 neutralization. This manuscript describes the nonclinical safety program supporting approval of the IL-
102
23p19 antagonist monoclonal antibody (mAb) tildrakizumab (Ilumya™ [tildrakizumab] full
103
prescribing information, 2018), also known as MK-3222, for the treatment of adults with
104
moderate to severe plaque psoriasis who are candidates for systemic therapy or phototherapy.
5
105
Tildrakizumab is a high-affinity (KD – 297 pM; as measured by surface plasmon resonance),
106
humanized IgG1/κ recombinant neutralizing mAb that selectively binds to the IL-23p19 subunit
107
and neutralizes human IL-23 (Kopp et al., 2015). The general expression pattern of IL-23 and
108
its cellular receptor is similar between human, cynomolgus monkey, and mouse; however, the
109
highest IL-23p19 amino acid identity occurs between human and cynomolgus monkey (98%).
110
The safety profile of tildrakizumab was characterized in an extensive nonclinical program
111
conducted in accordance with regulatory guidelines applicable to biotechnology products (ICH
112
M3[R2], 2009; US FDA, 1997) and international regulatory agency feedback throughout
113
development. Using a pharmacologically relevant cynomolgus monkey model system, the
114
toxicity profile was assessed in repeated-dose toxicity studies of up to 9 months in duration.
115
Potential developmental and reproductive effects were characterized in an embryo-fetal
116
development (EFD) study and a pre- and postnatal development (PPND) study in the
117
cynomolgus monkey.
118 119
2. MATERIALS AND METHODS
120
2.1 Ethics statements
121
All study protocols and animal housing were approved by the Institutional Animal Care
122
and Use Committee prior to study initiation, and animals were cared for in accordance with the
123
principles defined in the Guide for the Care and Use of Laboratory Animals (Institute for
124
Laboratory Animal Resources, 2011). All studies were conducted according to site Standard
125
Operating Procedures and the Organization for Economic Co-operation and Development
126
Principles of Good Laboratory Practice in the US.
127
6
128 129
2.2 Physical, chemical, and pharmaceutical properties Tildrakizumab was produced in Chinese Hamster Ovary cells using conventional cell
130
culture and purification processes. The parental mAb of tildrakizumab was produced by
131
immunization of IL 23p19-knockout mice with a recombinant chimeric IL-23 (human p19:
132
mouse p40) as the immunogen. The humanized anti–IL-23 antibody was obtained by grafting
133
the complementarity determining regions from the parental mAb onto the human IgG1
134
framework. Tildrakizumab (50 or 100 mg/mL) was supplied in a frozen liquid formulation and
135
stored at 2℃ to 8℃.
136 137 138
2.3 Animals and husbandry Chinese-origin cynomolgus monkeys (Macaca fascicularis) were housed at an Association
139
for Assessment and Accreditation of Laboratory Animal Care International-accredited facility in
140
species-specific indoor housing. Certified primate diet was provided daily in amounts
141
appropriate for the age and size of the animals, and tap water was available ad libitum to each
142
animal via an automatic watering device. Animals were provided enrichment by additional food
143
supplements and various cage-enrichment devices. Animals were maintained on a 12:12 hour
144
light:dark cycle at 18°C to 29°C with 30% to 70% relative humidity. All animals were negative
145
for simian retrovirus. The cynomolgus monkey was selected as an appropriate test species
146
based on sequence homology to human IL-23 and the similar picomolar binding
147 148
(KD ~ 50 pM) of tildrakizumab; no binding is observed in the rat and mouse. Tildrakizumab also has comparable cellular potency (~125 pM) to human and cynomolgus monkey IL-23.
149
7
150
2.4 Study design
151
2.4.1 Repeat-dose toxicity
152
Assessment of potential toxicity (with evaluation of functional endpoints for
153
cardiovascular, respiratory, and central nervous system [CNS] assessments) of tildrakizumab
154
was evaluated in cynomolgus monkeys in 2 repeat-dose studies of 3 and 9 months in duration.
155
Doses were selected to provide a mean anticipated exposure margin of 10-fold to the clinical
156
exposure and be associated with maximum pharmacology in monkeys based on internal cellular
157
potency. In the 3-month study, 4 groups of cynomolgus monkeys were administered either
158
placebo (Group 1), tildrakizumab 40 or 140 mg/kg by subcutaneous (SC) injection (Groups 2
159
and 3, respectively), or tildrakizumab 140 mg/kg by intravenous (IV) bolus (Group 4) every 2
160
weeks (Q2W) over 15 weeks. In the 9-month study, cynomolgus monkeys were administered a
161
SC dose of placebo or 10, 30, or 100 mg/kg tildrakizumab Q2W for 9 months. In both studies, a
162
4- to 6-month treatment-free postdose period was included. Necropsy was performed at the
163
end of dosing and after the treatment-free period; organ weights were measured, and tissues
164
were sampled for microscopic histopathology. During the study period, assessment of mortality,
165
clinical observations, and qualitative food consumption was made at least daily and body weight
166
was measured weekly. Physical examinations, electrocardiograms (ECG), blood pressure
167
measurements (via the use of cuffs), and ophthalmologic examinations were performed
168
predose, at multiple time points during the dosing period, and once during the treatment-free
169
period. Urine and blood were sampled for urinalysis, hematology, coagulation, and clinical
170
serum chemistry predose, at multiple time points during the dosing period, and once in the
171
treatment-free period. Urine was collected overnight, and standard urinalysis and urine
172
chemistry assessments were performed. Blood samples for toxicokinetic evaluation,
8
173
determination of IL-23 concentration, and immunogenicity analyses were collected as described
174
in the Supplemental Methods.
175
To ensure a sufficient number for fertility assessment, both the 3- and 9-month studies
176
contained sexually mature animals by study termination as determined by morphological
177
examination of the testes in males and daily vaginal swabs for evidence of menses and/or
178
hormone measurements in females. In the 9-month study, blood samples for estradiol analysis
179
were collected on multiple days of an individual female’s cycle and daily progesterone samples
180
were collected for 5 consecutive days during an individual female’s cycle.
181 182
2.4.2 Embryo-fetal development study
183
Pregnant cynomolgus monkeys were administered placebo or tildrakizumab SC at doses
184
of 10, 100, or 300 mg/kg once every 14 days from gestation day 20 (GD20) to GD118. Overall
185
health and food consumption were monitored daily, and pregnancy monitoring was performed
186
regularly via ultrasound to assess embryo-fetal loss (Tarantal and Hendrickx, 1988). Fetal
187
viability, gender, body weight, and placental weight were determined following Cesarean (C-)
188
section at GD140 ± 3 days. Blood samples for toxicokinetic evaluation and immunogenicity
189
analyses were collected as described in the Supplemental Methods. Following euthanasia,
190
external, visceral (including selected organ weights), and skeletal examinations were conducted
191
on all fetuses.
192 193 194
2.4.3 Pre- and postnatal development (PPND) study Pregnant cynomolgus monkeys were administered placebo or tildrakizumab 10 or
195
100 mg/kg SC once Q2W from GD50 until parturition. All dams were first-time mothers.
196
Maternal health and pregnancy progression were monitored as in the embryonic fetal
9
197
development study. Adult females were evaluated for changes in clinical signs, body weight,
198
and other parameters (including nursing behavior and milk let-down) for approximately 6
199
months postpartum. Milk samples were collected from mothers at postpartum day 28 (PPD28)
200
and PPD91 for toxicokinetic analysis. Blood samples for toxicokinetic and antidrug antibody
201
(ADA) analyses were collected from mothers at multiple gestation and postpartum days.
202
Clinical observations; body weight; nursing behavior; and external, morphometric, and
203
neurobehavioral assessments of cynomolgus monkey infants were collected for 6 months
204
postpartum. Blood samples from cynomolgus monkey infants were collected by venipuncture at
205
multiple time points for clinical pathology, toxicokinetic, ADA, and lymphocyte analyses. At
206
postnatal day (PND) 138 (± 2 days) and PND152 (± 2 days), cynomolgus monkey infants were
207
administered intramuscular keyhole limpet hemocyanin (KLH) as a vaccine surrogate. Blood was
208
collected via venipuncture before the first vaccination on PND138 and following the first
209
vaccination at multiple time points, before the second vaccination on PND152 and after the
210
second vaccination at multiple time points, and was analyzed by enzyme-linked immunosorbent
211
assay (ELISA) for the presence of anti-KLH IgG and anti-KLH IgM antibodies to assess T-cell
212
dependent antibody response (TDAR).
213
Mortality data during the study period were compared to the incidence of fetal and
214
neonatal losses in historical controls for untreated animals, as well as published literature of
215
spontaneous/background mortality rates among macaques (Small, 1982). Cynomolgus
216
monkey infants that died during the study period were examined for histomorphology. All
217
surviving cynomolgus monkey infants were euthanized on PND180 (± 2 days), and external and
218
visceral exams and histomorphology were conducted.
219
10
220 221
2.5 Immunophenotyping Quantification of cellular antigens and cell populations from cynomolgus monkey infant
222
blood samples and absolute counts of the subpopulations were generated using specific
223
antibodies against the marker antigens. Initial lymphocyte populations were identified and
224
gated for CD2+/CD20+ to identify all lymphocytes (sample purity/total B-cells.); CD20+ for B-
225
lymphocytes; CD3+ for T-lymphocytes; CD3+/CD4+ for T-helper lymphocytes, and CD3+/CD8+
226
for T-cytotoxic lymphocytes.
227 228
2.6 Bioanalytical, pharmacokinetic, toxicokinetic, pharmacodynamic, and anti-
229
tildrakizumab antibodies analyses
230
2.6.1 Assays for tildrakizumab in monkey serum and milk
231
A validated bridging electrochemiluminescence (ECL) assay using human IL-23 as
232
capture (dynamic range of 3.91–250 ng/mL) was used to quantify tildrakizumab in cynomolgus
233
monkey serum. An exploratory tildrakizumab ECL assay (dynamic range of 3.91–250 ng/mL)
234
was used to quantify tildrakizumab in cynomolgus monkey breast milk in the PPND study.
235 236 237
2.6.2 Assays for anti-tildrakizumab antibodies in monkey serum ADA formation in monkey serum was assessed in the 3- and 9-month repeat-dose
238
toxicity studies in monkeys with an ECL assay that was developed and validated using
239
biotinylated and ruthenylated tildrakizumab as the capture and detection reagents, respectively.
240
A protein G purified rabbit polyclonal (anti-tildrakizumab) antibody was used for the preparation
241
of the positive control. The assay had a dynamic range of 0.781 to 50 µg/mL expressed in
242
undiluted serum. ADA formation in serum samples from the EFD study was assessed using a
243
more sensitive ECL assay with a dynamic range of 100 (lower limit of quantitation [LLOQ]) to
11
244
12800 ng/mL (upper limit of quantitation). Based on the rabbit polyclonal positive control, the
245
sensitivity of this assay was 8.52 ng/mL. This ECL assay was subsequently transferred and
246
validated at Covance Laboratories, Inc. (Chantilly, VA, USA), and used to evaluate ADA in serum
247
samples derived from the monkey PPND study.
248 249 250
2.6.3 Assays for IL-23 in monkey serum In the 3-month toxicity study, IL-23 concentration in monkey serum was determined
251
using a validated ECL assay with an LLOQ of 250 pg/mL expressed in neat serum. The assay
252
used biotin- and SULFO-TAG™-labeled antihuman IL-23 mAbs that recognized an epitope of IL-
253
23 distinct from the one recognized by tildrakizumab. In the 9-month monkey toxicity study, a
254
validated assay with an LLOQ of 16 pg/mL expressed in neat serum was used. Both assays
255
were based on the Meso Scale Discovery platform.
256 257
2.6.4 Pharmacokinetic analysis
258
Serum concentration-time data were analyzed using model-independent methods
259
(Gibaldi and Perrier, 1982). Parameters determined included concentration extrapolated to time
260
0 (Co), maximum observed serum concentration (Cmax), time of maximum observed serum
261
concentration (Tmax), area under the concentration-time curve (AUC), area under the
262
concentration-vs-time curve from zero to time t (AUCt), systemic exposure (AUC[0–∞]),
263
bioavailability (F), accumulation ratio (R), clearance (CL), half-life (t1/2), and steady-state
264
volume of distribution (Vdss).
265
12
266 267
2.6.5 Computer software Pharsight® Knowledgebase Server™ versions 2.0.1 or 4.03 with WinNonlin version 4.0.1
268
(Pharsight Corporation, Cary, NC, USA) or Phoenix Application Suite version 1.3 (Pharsight
269
Corporation, Mountain View, CA, USA) was used to conduct the toxicokinetic analysis. Additional
270
analysis used Microsoft Excel (Redmond, WA, USA).
271 272
3. RESULTS
273
3.1 Chronic toxicity
274
In the 3- and 9-month repeat-dose toxicity studies, exposure to tildrakizumab was
275
independent of gender, increased with increasing dose, and increased with repeated IV or SC
276
administration (Tables 1 and 2). A low incidence (1 of 72 animals) of ADA response was noted
277
in the 3- and 9-month studies with an apparent increase in elimination rate of tildrakizumab
278
(data not shown). In the 9-month study, increases in total serum IL-23 concentrations were
279
observed in monkeys, consistent with binding of tildrakizumab with its target cytokine
280
(Supplemental Figure 1).
281
There was no mortality or effects on clinical observations, body weight, qualitative food
282
consumption, veterinary and ophthalmic evaluations, clinical chemistry, hematology, or
283
urinalysis parameters (data not shown). No toxicological findings were seen in assessment of
284
cardiovascular (including blood pressure and ECGs), respiratory, or CNS functions of
285
cynomolgus monkeys treated with tildrakizumab Q2W for up to 9 months (data not shown). In
286
addition, there were no tildrakizumab-related anatomic pathology (including macroscopic and
287
microscopic observations and organ weights). The no observed adverse effects level (NOAEL)
288
was 140 mg/kg (IV and SC) in the 3-month study and 100 mg/kg (SC) in the 9-month study.
289
13
290
3.2 Developmental and reproductive toxicity (DART)
291
3.2.1 Fertility
292
Communication of potential effects on fertility of new medicines is required in product
293
labels. When nonhuman primates are the only relevant toxicology species, the potential for
294
effects on male and female fertility can be evaluated by examination of reproductive tract
295
(organ weights and histomorphologic evaluation) within repeated-dose toxicity studies using
296
sexually mature monkeys. Therefore, both the 3- and 9-month studies included sexually mature
297
animals to determine potential effects of tildrakizumab on the testes histomorphology and/or
298
testicular weight in males; and vaginal swabs, evidence of menses, and/or hormone
299
measurements (estradiol and progesterone) in females. No tildrakizumab-related effects were
300
observed in any parameter evaluated in these studies.
301 302
3.2.2 Embryo-fetal development
303
In the EFD study, peak tildrakizumab serum concentrations were observed in maternal
304
animals from 1 to 7 days after the final dose on GD118 (Supplemental Figure 2). Systemic
305
maternal exposure to tildrakizumab increased with increasing dose (Table 3). Tildrakizumab
306
was detected in fetal serum at the time of C-section on GD140 (Table 5), indicating transfer
307
to the fetus (0.6–0.8 fetal/dam serum ratio). Concentrations of tildrakizumab in the fetus
308
ranged from 21.2 to 69.6, 0.0438 to 458, and 424 to 2180 µg/mL for the 10, 100, and 300
309
mg/kg groups, respectively. Although anti-tildrakizumab antibodies were detected in several
310
animals, including 1 fetus, there was no impact on the animals’ respective serum
311
concentration-time profile (Supplemental Figure 2), with the exception of 1 maternal
312
animal and fetus administered tildrakizumab 100 mg/kg. Anti-tildrakizumab antibodies were
313
present at C-section on GD140 in this dam and fetus; these samples were also positive for
14
314
neutralizing anti-tildrakizumab antibodies. This is consistent with the lower serum drug
315
concentration profile in the dam and fetus as compared with all other animals in the same
316
group.
317
There were no tildrakizumab-related effects on antemortem or postmortem
318
parameters measured, and there were no tildrakizumab-related abnormalities or variations
319
observed in any fetus, including fetal and placental weight at C-section (data not shown). In
320
addition, no toxicity was observed in dams in either the presence or absence of neutralizing
321
anti-tildrakizumab antibodies. The NOAEL was ≥300 mg/kg based on a lack of maternal or
322
developmental toxicity at any dose level.
323 324
3.2.3 Prenatal and postnatal development, including maternal function
325
Systemic exposure to tildrakizumab increased with increasing dose and repeated
326
administration in maternal animals (Table 4); in cynomolgus monkey infants, exposure
327
increased with increasing dose administration to maternal animals (Table 5). Ratios of mean
328
tildrakizumab serum concentrations in cynomolgus monkey infants to dams were
329
approximately 1 between PND7 and PND28 and increased up to 5 on PND180; cynomolgus
330
monkey infants appeared to have slightly reduced clearance relative to the dams from PPD7
331
to PPD180 based on the cynomolgus monkey infant/adult serum concentration ratio (Table
332
5). The mean postbirth serum tildrakizumab concentrations in the cynomolgus monkey infants
333
on PND180 were approximately 0.07 and 0.6 µg/mL at maternal doses of 10 and 100 mg/kg,
334
respectively. Tildrakizumab excretion in breast milk was minimal; mean tildrakizumab
335
concentrations in milk were approximately 0.09% to 0.2% of that in serum on PPD91 for 10 and
336
100 mg/kg, respectively. Some maternal animals in both dosing groups had anti-tildrakizumab
15
337
antibodies, and a subset of these had an apparent increase in tildrakizumab elimination rate
338
(Supplemental Figure 3).
339
Administration of tildrakizumab 10 and 100 mg/kg Q2W to pregnant cynomolgus
340
monkeys was well tolerated. There were no tildrakizumab-related changes in clinical signs,
341
food consumption, or body weights or effects on gestational length or pregnancy/postpartum
342
outcomes that were considered related to maternal administration of tildrakizumab (data not
343
shown).
344
Fetal loss rates (including abortions and stillbirths) were 20% (3/15) in the placebo
345
and 10 mg/kg groups, and 7% (1/15) in the 100 mg/kg group, and all occurred in the third
346
trimester (Table 6). Fetal loss incidence was within the historical control range for the testing
347
facility and in line with published data (Hendrie et al., 1996). Of the fetuses that were aborted
348
or stillborn in all groups, there were no tildrakizumab-related effects observed in the placenta
349
or umbilical cord (when available) or on body weights, morphometric measurements, external
350
or gross/visceral evaluations, or organ weights (data not shown).
351
The number of viable offspring delivered by natural birth was 11 in the placebo group,
352
12 in the 10 mg/kg tildrakizumab group, and 13 in the 100 mg/kg tildrakizumab group. Two
353
neonates (1 in placebo and 1 in the 100 mg/kg tildrakizumab group) were delivered by C-
354
section, resulting in a total of 12, 12, and 14 viable offspring in the control, 10 mg/kg, and
355
100 mg/kg groups, respectively. Of these viable offspring, 7 neonates died or were
356
euthanized within 15 days of birth and were examined by necropsy and histomorphology.
357
Neonatal loss clearly related to maternal neglect occurred in 5 cases within the first week of
358
postnatal life (Table 6); 1/12 (8%) in the placebo group, 2/12 (17%) in the 10 mg/kg group,
359
and 2/14 (14%) in the 100 mg/kg group. In the 10 mg/kg group, 1 moribund neonate was
360
sacrificed because it was unable to adequately grasp the dam to nurse, and another death
16
361
resulted from unsuccessful adoption following maternal death due to extensive blood loss
362
during delivery. Three other neonates were euthanized between PND1 and PND4 due to
363
maternal neglect (1 in the placebo group and 2 in the 100 mg/kg group); histopathological
364
examination showed slight depletion of lymphoid follicles in the spleen and/or moderate
365
lymphoid depletion in the thymus in these animals.
366
The incidence of neonatal loss determined not related to maternal neglect was 0% in
367
the placebo control group, 0% in the tildrakizumab 10 mg/kg group, and 14% (2/14) in the
368
tildrakizumab 100 mg/kg group (Table 6). These values are within the historical control
369
range of the testing facility for untreated animals. The 2 neonates from the 100 mg/kg group
370
that died on PND12 and PND15 appeared icteric with histological findings in the liver and
371
kidneys, suggestive of a possible viral infection.
372
Cynomolgus monkey infants surviving through 6 months postnatal to scheduled
373
necropsy (11, 10, and 10 in the placebo, 10 mg/kg, and 100 mg/kg groups, respectively)
374
showed no tildrakizumab-related changes in clinical signs; body weights; morphometric
375
measurements; external, heart, or gross/visceral assessments; neurobehavioral evaluations
376
(data not shown); clinical serum chemistry; hematology; lymphocyte phenotyping evaluation;
377
immune function competence TDAR test; organ weights; or anatomic pathology, including gross
378
and histopathological assessments (data not shown). In addition, no toxicity was observed in
379
the presence of neutralizing anti-tildrakizumab antibodies.
380
In summary, the no observed effects level (NOEL)/NOAEL in the PPND study was
381
established at 10 mg/kg as there was no difference between the incidences of fetal or neonatal
382
losses (due to maternal neglect or unrelated to maternal neglect) in this group compared to
383
historical controls. At 100 mg/kg, the 2 neonatal deaths due to potential viral infection were
384
considered of uncertain relationship to treatment with tildrakizumab based on the lack of
17
385
historical control histopathology data for occurrence of viral infection in neonate cynomolgus
386
monkeys.
387 388 389
3.3 Exposure multiples Systemic exposure (AUC) multiples for the 3- and 9-month repeat-dose toxicity, EFD,
390
and PPND cynomolgus monkey studies were calculated using clinical exposure derived from a
391
population pharmacokinetic model (AUC0-12 week of 305 µg·day/mL) at the recommended human
392
dose of 100 mg (Jauslin et al., 2019; Jauslin et al., 2018). The monkey-to-human exposure
393
multiple ranged from 7 to 159 times the recommended human dose at the NOAEL in these
394
studies (Table 7).
395 396 397
4. DISCUSSION AND CONCLUSIONS The safety profile of tildrakizumab was characterized in extensive nonclinical safety
398
studies. Tildrakizumab was well tolerated with no toxicological findings in cynomolgus
399
monkeys chronically treated with SC injections of tildrakizumab at 100 mg/kg for up to
400
9 months to maintain high systemic exposure.
401
The repeat-dose toxicity studies in cynomolgus monkeys also included the evaluation
402
of potential impact of tildrakizumab on cardiovascular, respiratory, and central nervous
403
system functions. No tildrakizumab-related changes in any measured safety pharmacology
404
endpoints were detected. These results in monkey studies are consistent with reports that IL-
405
23p19 null mutant mice are viable and breed normally, implying no overt effects in the
406
cardiovascular or other vital systems (Cua et al., 2003; Kurtz et al., 2014).
407 408
Similarly, in extensive developmental and reproductive toxicity studies in cynomolgus monkeys, tildrakizumab was well tolerated during pregnancy and no embryo-fetal toxicity was
18
409
observed in fetuses from pregnant monkeys administered tildrakizumab up to 300 mg/kg SC
410
every other week. In an additional PPND study in cynomolgus monkeys, there were no
411
tildrakizumab-related increases in fetal loss, neonatal loss, or combined fetal and neonatal
412
loss rates, and no effects on morphological or immunological development were observed in
413
offspring exposed to the drug in utero when pregnant monkeys were administered
414
tildrakizumab up to 100 mg/kg SC every other week.
415
As commonly seen in monkey PPND studies (Jarvis et al., 2010), 5 spontaneous
416
neonatal deaths were observed in the PPND study of tildrakizumab and determined to be
417
incidental and associated with maternal neglect. This is a common finding in cynomolgus
418
macaques, particularly in unexperienced, first-time pregnant dams who are less competent
419
than those who have previously given birth (Maestripieri et al., 1997; Prescott et al., 2012;
420
Tsuchida et al., 2008). Maternal neglect and failure to provide food are inducers of stress
421
(Dettling et al., 2007), which is consistent with the observed histopathology of lymphoid
422
tissues from neonates in the PPND study with tildrakizumab (Everds et al., 2013). Of these 5
423
deaths due to maternal neglect, 1 was also associated with failed adoption by a foster
424
mother. Nonhuman primate literature suggests that cross-fostering is less successful than
425
maternal-offspring bonding (Maestripieri, 2001).
426
In the tildrakizumab 100 mg/kg group, 2 additional neonatal deaths were attributed to
427
possible viral infection based on histopathology examination. However, there is a lack of
428
histological control data in nonhuman primate neonates, as neonates found dead typically do
429
not undergo necropsy and histopathology evaluation. Therefore, it is unknown if the observed
430
potential viral infection could be background finding or a drug-related effect. It seems unlikely
431
these observations are a drug-related effect given their low incidence, as well as the good
432
health and confirmed immune competence (ie, normal antibody response to KLH vaccination in
19
433
the TDAR test) of cynomolgus monkey infants monitored through 6 months after birth. Reports
434
of early perinatal and neonatal mortality (PND1−30) caused by infections, such as sepsis,
435
enteritis, and meningitis, in untreated nonhuman primates suggest these 2 neonatal deaths
436
may be coincidental (Price et al., 1973). Furthermore, selective inhibition of the IL-23p19
437
pathway is associated with decreased infection liability compared with broader IL-12/23p40
438
neutralization (Belladonna et al., 2006; Chackerian et al., 2006; Held et al., 2008; Kapil et al.,
439
2009; Khader et al., 2005; Kortylewski et al., 2009; Langowski et al., 2006; Rudner et al.,
440
2007; Schulz et al., 2008; Teng et al., 2010; Teng et al., 2012; Teng et al., 2011; von Scheidt
441
et al., 2014; Wu et al., 2007; Wu et al., 2009; Zelante et al., 2007). Nevertheless, a potential
442
relationship between these 2 neonatal deaths and the treatment of pregnant monkeys with
443
tildrakizumab cannot be ruled out.
444
Immune modulation is sometimes, but not always, associated with a potential for
445
carcinogenicity (Bongartz et al., 2006; Teng et al., 2015). Direct evaluation of carcinogenic
446
potential is not feasible in routine mouse or rat bioassays as tildrakizumab does not bind
447
murine IL-23, most likely due to lower sequence homology. However, multiple literature
448
reports suggest that treatment with a selective IL-23p19 blockade is not associated with an
449
increased tumor risk (Langowski et al., 2006; Teng et al., 2010; Teng et al., 2012; Teng et
450
al., 2011; von Scheidt et al., 2014).
451
In summary, the results of this comprehensive nonclinical safety program evaluating
452
tildrakizumab in cynomolgus monkeys support the safe use of tildrakizumab for the treatment
453
of psoriasis (Ilumya™ [tildrakizumab] full prescribing information, 2018).
454
20
455
5. ACKNOWLEDGEMENTS
456
The authors thank Drs. Kerry Blanchard, Eddie Bowman, Britta Mattson, Judith S. Prescott,
457
Sean Troth, and Gordon K. Wollenberg for their review of this manuscript; Melinda Marian and
458
Robert Venenziale for their roles in planning the early stages of the pharmacokinetic program;
459
and Megan Vogler for her technical input. Editorial support was provided by Kathleen Pieper,
460
PhD, of AlphaBioCom, LLC, and funded by Sun Pharmaceutical Industries, Inc.
461 462
6. AUTHOR CONTRIBUTIONS
463
All authors contributed to the conception, design, interpretation, critical review, and approval of
464
the manuscript to ensure data integrity and accuracy.
465 466
7. DECLARATION OF CONFLICTING INTERESTS
467
All authors are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc.,
468
Kenilworth, NJ, USA.
469 470
8. FUNDING
471
This work was funded by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc.,
472
Kenilworth, NJ, USA, and was conducted in the course of the authors’ employment.
21
473
9. REFERENCES
474
Abdollahi, E., et al., 2016. Protective role of R381Q (rs11209026) polymorphism in IL-23R gene
475
in immune-mediated diseases: A comprehensive review. J Immunotoxicol. 13, 286-300,
476
DOI:10.3109/1547691X.2015.1115448.
477 478 479
Belladonna, M. L., et al., 2006. IL-23 neutralization protects mice from Gram-negative endotoxic shock. Cytokine. 34, 161-9, DOI:10.1016/j.cyto.2006.04.011. Bongartz, T., et al., 2006. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of
480
serious infections and malignancies: systematic review and meta-analysis of rare
481
harmful effects in randomized controlled trials. JAMA. 295, 2275-85,
482
DOI:10.1001/jama.295.19.2275.
483
Boutet, M. A., et al., 2018. Role of the IL-23/IL-17 Axis in Psoriasis and Psoriatic Arthritis: The
484
Clinical Importance of Its Divergence in Skin and Joints. Int J Mol Sci. 19,
485
DOI:10.3390/ijms19020530.
486
Capon, F., et al., 2007. Sequence variants in the genes for the interleukin-23 receptor (IL23R)
487
and its ligand (IL12B) confer protection against psoriasis. Hum Genet. 122, 201-6,
488
DOI:10.1007/s00439-007-0397-0.
489
Chackerian, A. A., et al., 2006. Neutralization or absence of the interleukin-23 pathway does not
490
compromise immunity to mycobacterial infection. Infect Immun. 74, 6092-9,
491
DOI:10.1128/IAI.00621-06.
492 493 494 495
Cosentyx® [secukinumab] full prescribing information, 2018. Novartis Pharmaceutical Corporation. East Hanover, NJ. Cua, D. J., et al., 2003. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 421, 744-8, DOI:10.1038/nature01355.
22
496
Davies, R., et al., 2013. Influence of anti-TNF patient warning regarding avoidance of high risk
497
foods on rates of listeria and salmonella infections in the UK. Ann Rheum Dis. 72, 461-2,
498
DOI:10.1136/annrheumdis-2012-202228.
499
Dettling, A. C., et al., 2007. Behavioral and physiological effects of an infant-neglect
500
manipulation in a bi-parental, twinning primate: impact is dependent on familial factors.
501
Psychoneuroendocrinology. 32, 331-49, DOI:10.1016/j.psyneuen.2007.01.005.
502
Di Meglio, P., et al., 2011. The IL23R R381Q gene variant protects against immune-mediated
503
diseases by impairing IL-23-induced Th17 effector response in humans. PLoS One. 6,
504
e17160, DOI:10.1371/journal.pone.0017160.
505
Enbrel® [etanercept]. Full prescribing information, 2017. Amgen. Thousand Oaks, CA. .
506
Fredriksson, T., Pettersson, U., 1978. Severe psoriasis--oral therapy with a new retinoid.
507
Dermatologica. 157, 238-44.
508
Gibaldi, M., Perrier, D., 1982. Pharmacokinetics. 2nd ed Marcel Dekker, Inc., New York.
509
Held, K. S., et al., 2008. Generation of a protective T-cell response following coronavirus
510
infection of the central nervous system is not dependent on IL-12/23 signaling. Viral
511
Immunol. 21, 173-88, DOI:10.1089/vim.2008.0014.
512 513
Hendrie, T. A., et al., 1996. Frequency of prenatal loss in a macaque breeding colony. American Journal of Primatology. 40, 41-53.
514
Humira® [adalimumab] full prescribing information, 2018. AbbVie, Inc. North Chicago, IL.
515
ICH M3[R2], 2009. International Conference on Harmonisation (ICH) Tripartite Guidelines.
516
Guidance on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and
517
Marketing Authorization for Pharmaceuticals.
518 519
Ilumya™ [tildrakizumab] full prescribing information, 2018. Merck & Co, Inc. Whitehouse Station, NJ.
23
520 521 522
Institute for Laboratory Animal Resources, 2011. Guide for the Care and Use of Laboratory Animals, 8th ed. National Academic Press, Washington, DC. Jarvis, P., et al., 2010. The cynomolgus monkey as a model for developmental toxicity studies:
523
variability of pregnancy losses, statistical power estimates, and group size
524
considerations. Birth Defects Res B Dev Reprod Toxicol. 89, 175-87,
525
DOI:10.1002/bdrb.20234.
526
Jauslin, P., et al., 2019. Population-Pharmacokinetic Modeling of Tildrakizumab (MK-3222), an
527
Anti-Interleukin-23-p19 Monoclonal Antibody, in Healthy Volunteers and Subjects with
528
Psoriasis. Clin Pharmacokinet. DOI:10.1007/s40262-019-00743-7.
529
Jauslin, P., et al., 2018. Abstract 6721. Population pharmacokinetic modeling of tildrakizumab
530
(MK-3222), an anti-interleukin-23p19 monoclonal antibody, in healthy volunteers and
531
subjects with psoriasis. J Am Acad Dermatol. 79, AB224.
532
Kapil, P., et al., 2009. Interleukin-12 (IL-12), but not IL-23, deficiency ameliorates viral
533
encephalitis without affecting viral control. J Virol. 83, 5978-86, DOI:10.1128/JVI.00315-
534
09.
535
Khader, S. A., et al., 2005. IL-23 compensates for the absence of IL-12p70 and is essential for
536
the IL-17 response during tuberculosis but is dispensable for protection and antigen-
537
specific IFN-gamma responses if IL-12p70 is available. J Immunol. 175, 788-95.
538
Kopp, T., et al., 2015. Clinical improvement in psoriasis with specific targeting of interleukin-23.
539 540 541
Nature. 521, 222-6, DOI:10.1038/nature14175. Kortylewski, M., et al., 2009. Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. Cancer Cell. 15, 114-23, DOI:10.1016/j.ccr.2008.12.018.
24
542
Kubota, K., et al., 2015. Epidemiology of psoriasis and palmoplantar pustulosis: a nationwide
543
study using the Japanese national claims database. BMJ Open. 5, e006450,
544
DOI:10.1136/bmjopen-2014-006450.
545
Kurtz, S. L., et al., 2014. IL-23 p19 knockout mice exhibit minimal defects in responses to
546
primary and secondary infection with Francisella tularensis LVS. PLoS One. 9, e109898,
547
DOI:10.1371/journal.pone.0109898.
548
Langley, R. G., et al., 2013. Safety results from a pooled analysis of randomized, controlled
549
phase II and III clinical trials and interim data from an open-label extension trial of the
550
interleukin-12/23 monoclonal antibody, briakinumab, in moderate to severe psoriasis. J
551
Eur Acad Dermatol Venereol. 27, 1252-61, DOI:10.1111/j.1468-3083.2012.04705.x.
552 553 554
Langowski, J. L., et al., 2006. IL-23 promotes tumour incidence and growth. Nature. 442, 4615, DOI:10.1038/nature04808. Langrish, C. L., et al., 2005. IL-23 drives a pathogenic T cell population that induces
555
autoimmune inflammation. J Exp Med. 201, 233-40, DOI:10.1084/jem.20041257.
556
Maestripieri, D., 2001. Is there mother–infant bonding in primates? Developmental Review. 21,
557 558 559 560
93-120. Maestripieri, D., et al., 1997. Infant abuse runs in families of group-living pigtail macaques. Child Abuse Negl. 21, 465-71. Mease, P. J., 2011. Measures of psoriatic arthritis: Tender and Swollen Joint Assessment,
561
Psoriasis Area and Severity Index (PASI), Nail Psoriasis Severity Index (NAPSI), Modified
562
Nail Psoriasis Severity Index (mNAPSI), Mander/Newcastle Enthesitis Index (MEI), Leeds
563
Enthesitis Index (LEI), Spondyloarthritis Research Consortium of Canada (SPARCC),
564
Maastricht Ankylosing Spondylitis Enthesis Score (MASES), Leeds Dactylitis Index (LDI),
565
Patient Global for Psoriatic Arthritis, Dermatology Life Quality Index (DLQI), Psoriatic
25
566
Arthritis Quality of Life (PsAQOL), Functional Assessment of Chronic Illness Therapy-
567
Fatigue (FACIT-F), Psoriatic Arthritis Response Criteria (PsARC), Psoriatic Arthritis Joint
568
Activity Index (PsAJAI), Disease Activity in Psoriatic Arthritis (DAPSA), and Composite
569
Psoriatic Disease Activity Index (CPDAI). Arthritis Care Res (Hoboken). 63 Suppl 11,
570
S64-85, DOI:10.1002/acr.20577.
571 572 573 574
Prescott, M. J., et al., 2012. Laboratory macaques: When to wean? Applied Animal Behaviour Science. 137, 194-207. Price, R., et al., 1973. Simian neonatology: III. The causes of neonatal mortality. Veterinary Pathology. 10, 37-44.
575
Remicade® [infliximab] full prescribing information, 2018. Jassen Biotech, Inc. Horsham, PA.
576
Rudner, X. L., et al., 2007. Interleukin-23 (IL-23)-IL-17 cytokine axis in murine Pneumocystis
577 578
carinii infection. Infect Immun. 75, 3055-61, DOI:10.1128/IAI.01329-06. Schulz, S. M., et al., 2008. Protective immunity to systemic infection with attenuated Salmonella
579
enterica serovar enteritidis in the absence of IL-12 is associated with IL-23-dependent
580
IL-22, but not IL-17. J Immunol. 181, 7891-901.
581 582 583 584
Siliq™ [broadalumab] full prescribing information, 2017. Valeant Pharmaceuticals North America, LLC. Bridgewater, NJ. Small, M. F., 1982. Reproductive failure in macaques. American Journal of Primatology. 2, 137147.
585
Stelara® [ustekinumab] full prescribing information, 2018. Janssen Biotech, Inc. Horsham, PA.
586
Taltz® [ixekizumab] full prescribing information, 2018. Eli Lilly and Compnay. Indianapolis, IN. .
587
Tarantal, A. F., Hendrickx, A. G., 1988. Use of ultrasound for early pregnancy detection in the
588
rhesus and cynomolgus macaque (Macaca mulatta and Macaca fascicularis). J Med
589
Primatol. 17, 105-12.
26
590
Teng, M. W., et al., 2010. IL-23 suppresses innate immune response independently of IL-17A
591
during carcinogenesis and metastasis. Proc Natl Acad Sci U S A. 107, 8328-33,
592
DOI:10.1073/pnas.1003251107.
593
Teng, M. W., et al., 2015. IL-12 and IL-23 cytokines: from discovery to targeted therapies for
594
immune-mediated inflammatory diseases. Nat Med. 21, 719-29, DOI:10.1038/nm.3895.
595
Teng, M. W., et al., 2012. Opposing roles for IL-23 and IL-12 in maintaining occult cancer in an
596 597
equilibrium state. Cancer Res. 72, 3987-96, DOI:10.1158/0008-5472.CAN-12-1337. Teng, M. W., et al., 2011. Anti-IL-23 monoclonal antibody synergizes in combination with
598
targeted therapies or IL-2 to suppress tumor growth and metastases. Cancer Res. 71,
599
2077-86, DOI:10.1158/0008-5472.CAN-10-3994.
600 601
Tonel, G., et al., 2010. Cutting edge: A critical functional role for IL-23 in psoriasis. J Immunol. 185, 5688-91, DOI:10.4049/jimmunol.1001538.
602
Tremfya® [guselkumab] full prescribing information, 2017. Jassen Biotech, Inc. Hrosham, PA.
603
Tsuchida, J., et al., 2008. Maternal behavior of laboratory-born, individually reared long-tailed
604 605
macaques (Macaca fascicularis). J Am Assoc Lab Anim Sci. 47, 29-34. US FDA, 1997. Points to Consider the Manufacture and Testing of Monoclonal Antibody Products
606
for Human Use. US Department of Health and Human Services, Center for Biologics
607
Evaluation and Research.
608
von Scheidt, B., et al., 2014. Combined anti-CD40 and anti-IL-23 monoclonal antibody therapy
609
effectively suppresses tumor growth and metastases. Cancer Res. 74, 2412-21,
610
DOI:10.1158/0008-5472.CAN-13-1646.
611
World Health Organization, Global Report on Psoriasis. 2016.
612
http://apps.who.int/iris/bitstream/10665/204417/1/9789241565189_eng.pdf Accessed
613
Jan 8, 2019.
27
614
Wu, Q., et al., 2007. IL-23-dependent IL-17 production is essential in neutrophil recruitment
615
and activity in mouse lung defense against respiratory Mycoplasma pneumoniae
616
infection. Microbes Infect. 9, 78-86, DOI:10.1016/j.micinf.2006.10.012.
617 618 619
Wu, S., et al., 2009. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med. 15, 1016-22, DOI:10.1038/nm.2015. Zelante, T., et al., 2007. IL-23 and the Th17 pathway promote inflammation and impair
620
antifungal immune resistance. Eur J Immunol. 37, 2695-706,
621
DOI:10.1002/eji.200737409.
28
622
10. TABLES AND FIGURES
623
Table 1. Mean toxicokinetic parameters in cynomolgus monkeys (sexes combined) following
624
administration of tildrakizumab in the 3-month repeat-dose toxicity study
Dose route IV
Number of dosesa
Sex (n)
1
F (6) M (6)
7
Nominal AUC(0-14 days) dose µg·day/mL)b (mg/kg) (µ 140
19300 (9)
Cmax (µ µg/mL)b
Tmax (days)c
Mean t½ (days)b,d
Re
3220 (18)
NA
NA
NA f
47200 (37)
6990 (30)
NA
17.7 (16)
2.44
40
6900 (29)
709 (40)
3 (1–14)
NA
NA
140
15900 (18)
1380 (22)
3 (1–14)
NA
NA
40
14100 (59)
1200 (62)
3 (1–7)
21.0 (17)
2.04
140 40500 (53) 3810 (58) 3 (0–3) 17.9 (17) Animals received a SC dose of 40 or 140 mg/kg or an IV dose of 140 mg/kg Q2W for 3
2.56
1
F (6) M (6)
7
F (6) M (6)
SC
625
a
626
months. Data is shown for animals receiving either the first or seventh Q2W dose.
627
b
628
c
629
administered as an IV bolus, as the timing of the bolus can affect the observed Tmax.
630
d
631
per sex per group). The mean elimination t½ was 18.9 days across groups (n = 17).
632
e
633
f
634
antibodies on tildrakizumab serum concentrations.
635
AUC, area under the serum concentration-time curve; Cmax, maximum observed serum
636
concentration; IV, intravenous; NA, not applicable; Q2W, once every 2 weeks; R, accumulation
637
ratio; SC, subcutaneous; t½, terminal phase half-life; Tmax, time of maximum observed serum
638
concentration.
Geometric means (coefficient of variation).
Median (minimum−maximum). Tmax is not listed for studies in which tildrakizumab was
Elimination t½ was determined for the animals assigned to the postdose period (n = 3 animals
Accumulation (R) = AUC(0–14 days)Dosing Interval n (after repeated dosing) ÷ AUC(0–14 days)Dosing Interval 1.
One monkey was excluded from t½ calculations due to the influence from anti-tildrakizumab
29
639
Table 2. Mean toxicokinetic parameters in cynomolgus monkeys (sexes combined) following SC
640
administration of tildrakizumab in the 9-month repeat-dose toxicity study Number of dosesa
Nominal dose (mg/kg)
AUC(0-14 days) (µ µg·day/mL)b
Cmax (µ µg/mL)b
Tmax (day)c
Rd
10 30 100
1190 (50) 3340 (21) 11600 (25)
133 (97) 476 (30) 1080 (29)
1 (0.25–14) 1 (1–1) 3 (0.25–3)
NA NA NA
10 ≥13 30 100 a Animals were dosed Q2W.
3290 (29) 9250 (27) 27500 (39)
295 (31) 893 (29) 2640 (51)
3 (0.0833–7) 3 (1–3) 3 (1–7)
2.95 2.92 2.69
F (6) M (6)
1
e
641
Sex (N)
F (12) M (12)
642
b
643
c
644
d
645
e
646
12 animals/sex assessed).
647
AUC, area under the serum concentration-time curve; Cmax, maximum observed serum
648
concentration; NA, not applicable; Q2W, once every 2 weeks; R, accumulation ratio; SC,
649
subcutaneous; Tmax, time of maximum observed serum concentration.
Mean (coefficient of variation).
Median (range). Accumulation (R) = AUC(0–14 days)Dosing Interval n (after repeated dosing) ÷ AUC(0–14 days)Dosing Interval 1.
Mean of data from dosing intervals 13 and 19 (N = 6 animals/sex per interval for a total of
30
650
Table 3. Mean toxicokinetic parameters following subcutaneous administration of tildrakizumab
651
in pregnant cynomolgus monkeys on gestation day 118
Number of dosesa 8
Nb
Nominal dose (mg/kg)
AUC(0-14 days) (µ µg·day/mL)c
12 11 11
10 100 300
1720 (17) 16300 (27) 48500 (22)
Cmax (µ µg/mL)c 151 (20) 1600 (25) 4820 (21)
Tmax (day)d 3 (3–7) 3 (3–3) 3 (1–3)
652
a
653
b
654
c
655
d
656
AUC, area under the serum concentration-time curve; Cmax, maximum observed serum
657
concentration; GD, gestation day; Tmax, time of maximum observed serum concentration.
Monkeys were administered every 14 days from GD20 to GD118. Number of pregnant dams.
Mean (coefficient of variation). Median (range).
31
658
Table 4. Maternal serum tildrakizumab toxicokinetic parameters on gestation day 120 in the
659
pre- and postnatal development study
Day GD 120 660
Dosea (mg/kg/dose)
AUC0-336 hr (µg/mL·hr)
Cmax (µg/mL)
Tmax (hr)
10
47500 (2820)b
193 (14.7)
54 (5.4)
1760 (107)
61 (4.4)
c
100 434000 (28500) Data are presented as mean (SE).
661
a
662
b
663
after dose administration on GD134.
664
c
665
these animals, the sample at 336 hours postdose on GD120 was collected after dose
666
administration on GD134.
667
AUC, area under the serum concentration-time curve; Cmax, maximum observed serum
668
concentration; GD, gestation day; Q2W, once every 2 weeks; SE, standard error; Tmax, time of
669
maximum observed serum concentration.
Animals were dosed Q2W from GD50 until parturition. One animal was excluded because the sample at 336 hours postdose on GD120 was collected
Two animals were excluded due to insufficient quantifiable time points; additionally, in 1 of
32
670
Table 5. Cynomolgus monkey infant/adult female serum tildrakizumab concentration ratios
671
following tildrakizumab administration to adult females prebirth in the pre- and postnatal
672
development study
Daya 7 28 91 180 ± 2 673
Doseb (mg/kg/dose)
Infant monkey serum concentration (µg/mL)
Adult female monkey serum concentration (µg/mL)
Infant monkey/adult female monkey ratio
10
49.3 (5.5)
51.2 (7.9)
1.0
100
466 (26.1)
468 (56.6)
1.0
10
22.7 (2.6)
19.7 (3.7)
1.2
100
213 (18.2)
157 (22.7)
1.4
10
2.53 (0.46)
0.86 (0.3)
2.9
100
21.9 (2.9)
8.3 (3.1)
2.6
10
0.075 (0.02)
0.015 (0.01)
4.9
0.60 (0.15)
0.28 (0.15)
2.2
100 Data are presented as mean (SE).
674
a
675
b
676
GD, gestation day; PND, postnatal day; PPD, postpartum day; SE, standard error.
PND for cynomolgus monkey infants and PPD for adult females. Maternal animals were dosed once every 2 weeks from GD50 until parturition.
33
677
Table 6. Summary of fetal and neonatal losses in pre- and postpartum developement study Treatment group (number of animals)
Fetal loss (abortion and stillbirths)
Neonatal loss due to maternal neglecta
Neonatal loss not due to maternal neglect
Combined fetal and neonatal loss
Placebo (N = 15)
3/15 (20)
1/12 (8)
0/12
4/15 (27)
Tildrakizumab at 10 mg/kg (N = 15)
3/15 (20)
2/12 (17)
0/12
5/15 (33)
Tildrakizumab at 100 mg/kg (N = 15)
1/15 (7)
2/14 (14a)
2/14 (14)
5/15 (33)
Historical controlsb
7%–29%
0%–17%
0%–20%
17%–43%
678
Data are presented as fetal or neonatal loss per total number of animals (%).
679
a
680
b
Includes 1 neonatal death at 100 mg/kg unable to nurse. Data on file.
34
681
Table 7. Systemic tildrakizumab exposure multiples Dose levels a (mg/kg/day) 40 140 (NOAEL) 140 via IV (NOAEL)
AUCt (µg·day/mL) 14100 40500 47200
Monkey to human b exposure multiple NA 133 155
9 months
10 30 100 (NOAEL)
3290 9250 27500
NA NA 90
EFD
10 100 300 (NOAEL)
1720 16300 48500
NA NA 159
10 1979c 100 (NOAEL) 18083c a Subcutaneous route of administration, except where noted.
7 59
Study in monkeys 3 months
PPND 682 683
b
Exposure multiples calculated from a population pharmacokinetic model derived from the
684
human AUC0-12week of 305 ug·day/mL for 100 mg (MRHD) (Jauslin et al., 2019; Jauslin et al.,
685
2018).
686
c
AUC (µg·hr/mL)/24 hours to convert to AUC (µg/day/mL).
687
AUCt, area under the concentration-vs-time curve from zero to time t; EFD, embryo-fetal
688
development; MRHD, recommended human dose; NA, not applicable; NOAEL, no observed
689
adverse effect level; PPND, pre- and postnatal development; IV, intravenous.
35
690
11. SUPPLEMENTAL MATERIAL
691
Supplemental Methods
692
Three-month repeat-dose toxicity study collection time points
693
Blood samples for toxicokinetic evaluation and determination of interleukin (IL)-23
694
concentration were collected prior to dosing on the first day and at multiple time points within
695
the first 24 hours postdose, then at 24-hour intervals up to 96 hours, and at 168 hours
696
following the first dose. Thereafter, blood samples for toxicokinetic evaluation, IL-23
697
concentration measures, and antidrug antibody (ADA) analyses were collected at day 10; prior
698
to dosing every 2 weeks on days 14 through 70; on days 98, 84, and 112; and every other
699
week after until sacrifice. On day 84, additional samples were taken for toxicokinetic analyses
700
predose and at multiple time points up to 168 hours postdose.
701 702 703
Nine-month repeat-dose toxicity study collection time points Blood samples for toxicokinetic evaluation were collected on days 1, 169, and 253 (at
704
predose and multiple time points up to 336 hours postdose on each of those days); predose on
705
days 43, 85, 127, and 211; at week 8 of the treatment-free period; and on the day of sacrifice.
706
Blood was collected for ADA analysis and IL-23 concentration measures at predose; on days 1,
707
15, 43, 85, 127, 169, and 211; week 8 of the treatment-free period; and on the day of sacrifice.
708
Additional samples for IL-23 concentration were taken on days 3 and 7.
709 710 711 712
Embryo-fetal development study collection time points Maternal blood was sampled predose on gestation day (GD)20, GD34, GD62, and GD90; at multiple time points up to 336 hours postdose on GD118; and once on the day of Cesarean-
36
713
section (GD140) for toxicokinetic and ADA analyses, including a neutralization assay. Fetal
714
umbilical cord blood samples were collected at C-section for toxicokinetic and ADA analyses.
37
715
Supplemental Figure 1. Individual serum IL-23 levels over time during the 9-month repeat-
716
dose toxicity study in males
717 718
N = 6 for all doses. Each line represents an individual male; female data was similar. Day 0 was
719
the first day of tildrakizumab administration. Day 318 and 395 measurements occurred during
720
the recovery phase.
721
a
722
increase in IL-23 in a single monkey at the day 169 time point is unknown but did not affect the
723
toxicity profile.
724
IL, interleukin.
Tildrakizumab exposure in the 30 mg/kg group showed low variability. The reason for the
725
38
726
Supplemental Figure 2. Maternal tildrakizumab serum concentration and antidrug antibody
727
presence during the embryofetal development study
728 729
a
730
b
731
concentrations were measured prior to dosing (trough concentration) with robust sampling after
732
the final dose.
733
Day 0 was the first day of drug administration. Each line represents an individual animal. Time
734
points where ADA was detected are marked with a red X. All dose groups, but not the placebo
735
group, are presented. One animal was positive on day 140 and 160; this animal and the animal
736
positive on day 34 were different from the 5 that were positive on day 20.
737
ADA, antidrug antibody; GD, gestation day; TIL, tildrakizumab.
Five animals were ADA-positive at day 20. Monkeys were administered every 14 days from GD20 to GD118. Tildrakizumab serum
738
39
Supplemental Figure 3. Maternal tildrakizumab serum concentration and antidrug antibody presence during the pre- and postnatal development study for A) the 10 mg/kg group and B) the 100 mg/kg group A)
a
Three animals were ADA-positive at GD50, and 5 animals were ADA-positive at postpartum day 91.
b
Animals were dosed Q2W from GD50 until parturition. Tildrakizumab serum concentrations were measured prior to dosing (trough
concentration) with robust sampling after the final dose. c
Four animals were ADA-positive on postpartum day 180; all animals except 1 that were ADA-positive on postpartum day 91 were
also ADA-positive on postpartum day 180.
40
Each line represents an individual maternal animal’s tildrakizumab serum concentration. Time points where ADA were detected are marked with a red X. ADA, antidrug antibody; GD, gestation day; Q2W, every 2 weeks; TIL, tildrakizumab.
B)
a
Three animals were ADA-positive at gestation day 50.
41
b
Animals were dosed Q2W from GD50 until parturition. Tildrakizumab serum concentrations were measured prior to dosing (trough
concentration) with robust sampling after the final dose. Each line represents an individual maternal animal’s tildrakizumab serum concentration. Time points where ADA were detected are marked with a red X. ADA, antidrug antibody; GD, gestation day; Q2W, every 2 weeks; TIL, tildrakizumab.
42
FUNDING This work was funded by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA and was conducted in the course of the authors’ employment.
Declaration of interests ☐ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. ☒The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:
All authors are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA.