Uploaded by 刘文禹

A comprehensive review of the pharmacokinetics of approved therapeutic monoclonal antibodies in Japan Are Japanese phase I studies still needed

advertisement
Review
A Comprehensive Review of the
Pharmacokinetics of Approved Therapeutic
Monoclonal Antibodies in Japan: Are Japanese
Phase I Studies Still Needed?
The Journal of Clinical Pharmacology
54(5) 483–494
© 2014, The American College of
Clinical Pharmacology
DOI: 10.1002/jcph.231
Koji Chiba, PhD1,2, Hiroyuki Yoshitsugu, PhD3, Yuto Kyosaka, BSc2,
Satofumi Iida, PhD4, Koichiro Yoneyama, MSc4, Takahiko Tanigawa, PhD5,
Takashi Fukushima, BSc2†, and Masaki Hiraoka, PhD6
Abstract
Ethnic evaluation of the pharmacokinetics and safety of new drugs is required in Japan before implementing bridging or joining global studies. As
therapeutic monoclonal antibodies (mAbs) show limited ethnic differences, their pharmacokinetics and safety in Japanese individuals could be estimated
from prior non‐Japanese studies. Therefore, there is potential to re‐evaluate the development program for mAbs in Japan. We reviewed the
pharmacokinetics of mAbs approved in Japan. Although some differences had been observed in pharmacokinetics of mAbs between Japanese and non‐
Japanese populations (mainly Caucasians), these differences were attributed to differences of body weight and/or antigen levels. Moreover, the influential
factors can be estimated without conducting regional pharmacokinetic/safety studies. The pharmacokinetics of some mAbs is presumably non‐linear and
show differences between healthy volunteers and patients because of differences in antigen levels. However, for 10/24 mAbs approved in Japan, Japanese
healthy volunteer studies were conducted before the patient studies. Additionally, for the mAbs that showed ethnic differences in pharmacokinetics, the
doses selected in subsequent patient studies were the same as the doses approved in the United States. In this review, we discuss new drug development
strategies in various regions, and assess the need for regional pharmacokinetics/safety studies before joining global studies.
Keywords
monoclonal antibody preparations, Japan, regulatory affairs
The clinical trial environment in Japan has matured over
the last decade, at least in part following the introduction
of the International Conference on Harmonization Guidelines for Ethnic Factors in the Acceptability of Foreign
Clinical Data (ICH‐E5, R1),1 and its adoption by the
Pharmaceutical and Medical Devices Agency (PMDA).2
This guideline proposed that the similarity of pharmacokinetics between Japanese and non‐Japanese individuals
should be evaluated before proceeding to bridging studies,
which could be performed in a new region to extrapolate
foreign clinical data to the population in the new region.
The objective of this bridging approach is to minimize
duplication of large clinical trials.
The clinical trial environment in Japan was further
revolutionized in 2007 by the recommendations of the
Ministry of Health, Labour and Welfare (MHLW) for
Japanese centers to participate in global clinical trials.3 As
a consequence of these recommendations, the number of
global clinical trials that have included Japanese clinical
sites has markedly increased.4
To assess the feasibility of joining global clinical trials,
possible ethnic differences between Japanese and non‐
Japanese subjects must be evaluated, as these may adversely
influence the safety and efficacy of the new drug. Therefore,
it is advocated that pharmacokinetic studies must be
conducted in Japanese subjects before implementing
bridging studies or joining global clinical trials.
1
Laboratory of Clinical Pharmacology, Yokohama College of Pharmacy,
Yokohama, Japan
2
Department of Drug Development and Regulatory Science, Keio
University Graduate School of Pharmaceutical Science, Tokyo, Japan
3
Discovery Medicine & Clinical Pharmacology, Research & Development, Bristol‐Myers Squibb, Princeton, NJ, USA
4
Clinical Research Planning Department, Chugai Pharmaceutical Co.,
Ltd., Tokyo, Japan
5
Clinical Sciences, Bayer Pharma AG, Berlin, Germany
6
DMCP, Clinical Research, Bristol‐Myers K.K., Tokyo, Japan
*Present address: Clinical Pharmacology & PPDM, Japan Development,
MSD K.K., Tokyo, Japan.
y
Present address: Site Management Department, Janssen
Pharmaceutical K.K., Tokyo, Japan.
Submitted for publication 26 September 2013; accepted 7 November
2013.
Corresponding Author:
Masaki Hiraoka, PhD, DMCP, Clinical Research, Bristol‐Myers K.K.,
Tokyo, Japan, Shinjuku i‐Land Tower 5‐1, Nishi‐Shinjuku 6‐chome,
Shinjuku‐ku, Tokyo 163‐1328, Japan
E‐mail: masaki.hiraoka@bms.com
Ethnic Differences in the
Pharmacokinetics of Monoclonal
Antibodies
Because of the large size, charge, polarity, and hydrophilicity of mAbs and the morphology of the paracellular
pores in the vascular endothelium, the distribution of mAbs
to peripheral tissue is limited and slow.5 Paracellular
movement is thought to be the main route by which mAbs
transfer from the vascular space to the interstitial space.6
Transcellular movement of mAbs by receptor‐mediated
endocytosis, phagocytosis, and fluid‐phase pinocytosis
mainly occurs in the vascular or interstitial space. Once the
mAbs are incorporated into the cells, some of them enter
the FcRn salvage pathway and are returned to the
interstitial or vascular space. They may also return to the
circulation via lymphatic drainage.7 Consequently, mAbs
are mainly distributed in blood and interstitial spaces. From
this biochemical perspective, mAbs are less likely to be
sensitive to extrinsic or intrinsic ethnic factors compared
with small chemical compounds.8,9 So far, there have been
no reports describing ethnic differences, at least, in the
paracellular movement of mAbs.
Figure 1 shows the differences in clearance routes
between large and small molecule drugs, including mAbs,
Patients
Internalization
CDC
ADCC
Efficacy
Toxicity
Membrane
associated
target
antigens
Soluble
target
antigensa
Catabolism
Target mediated
clearance
Healthy volunteers
Phagocytic
cells
FcRn
Blood
Enzyme
Receptor
Efficacy
Toxicity
Blood
Kidney
Toxicity
Other
organs
Toxicity
Liver
Toxicity
Hepatic clearance
Monoclonal antibodies
(Large molecule drugs)
It is also important to consider whether ethnic
differences in pharmacokinetics may translate into
clinically meaningful differences in efficacy or safety.
This issue raised the question of whether pharmacokinetic
studies are necessary in the early phase of development for
all drugs, or whether the need for such studies should be
considered on a case‐by‐case basis, considering the
characteristics of the target population (e.g., genetic
mutational status and relative expression in different
ethnic groups), as well as the expected absorption,
distribution, metabolism, and excretion of the drug.
Antibody drugs show several desirable characteristics,
including good solubility and stability, long presence in
the body, high selectivity and specificity, and low risk for
bioconversion to toxic metabolites. Consequently, numerous monoclonal antibodies (mAbs) have been approved as
therapeutic drugs and many mAbs are currently under
clinical evaluation. Therefore, it is predicted that many
mAbs will be introduced into the market.
The primary objective of this review was to examine
whether the characteristics of mAbs in non‐Japanese
subjects, mainly Caucasians, could predict their pharmacokinetic characteristics in Japanese subjects, and whether
a specific and abbreviated development strategy for mAbs
in Japan could be considered. We expect that this regional
investigation could be expanded and applied to Asian
regulatory science. We included mAbs and mAb‐like
therapeutic proteins, such as fusion proteins containing the
Fc domain of IgG, in this review.
The Journal of Clinical Pharmacology / Vol 54 No 5 (2014)
Small molecule drugs
484
Renal clearance
Figure 1. Clearance of small and large molecule drugs. The light blue‐
squared region represents the metabolism and disposition in healthy
volunteers and patients. The yellow region indicates patient‐specific
metabolism and disposition. The sections below and above the dotted
line represent small molecule drugs and large molecule drugs (mAbs),
respectively. Small molecule drugs are ubiquitously distributed and are
mainly metabolized in the liver or eliminated by the kidney. The efficacy
and toxicity of small molecule drugs are mediated by their interactions
with enzymes, receptors, and other proteins in target tissues and in other
organs. Large molecule drugs, including mAbs, are only distributed by the
blood and are eliminated by target‐mediated clearance or phagocytosis.
Because the target is highly expressed in patients, target‐mediated
clearance is the predominant clearance route in patients. ADCC,
antibody‐dependent cellular cytotoxicity; CDC, complement‐dependent
cytotoxicity; PK, pharmacokinetics. aSome antibody drugs bind to
endogenous substances.
in healthy volunteers and patients. In terms of the
metabolism of mAbs, the majority of the mAb dose is
cleared by intracellular catabolism, through Fc‐receptor‐
mediated clearance and target‐mediated clearance.6 mAbs
against soluble antigens are primarily eliminated by Fc
receptor‐mediated clearance, which involves a non‐
specific common pathway for endogenous IgG and
mAbs via FcRn and FcgR, a process that shows linear
pharmacokinetics. Several reports have described ethnic
differences in these pathways.10–13 For FcRn, genetic
polymorphisms of FCGRT were reported to show ethnic
differences, as the mean allelic frequency of the variable
number of tandem repeats was 0.032 in Japanese
individuals versus 0.075 in Caucasians.10 However, the
impact of these genetic variations on the function of FcRn
is yet to be established. For FcgR, although ethnic
differences were reported in the frequencies of variants of
FcgRIIa, FcgRIIb, FcgRIIIa, and FcgRIIIb,11–13 their
effects on the clearance of mAbs remain unknown.
The absence of ethnic differences of endogenous IgG in
Fc receptor‐mediated clearance indicates the absence of
ethnic differences in the elimination and clearance of
mAbs because of the involvement of non‐specific
common pathways. mAbs against antigens expressed on
cell surfaces are eliminated by Fc receptor‐mediated and
target‐mediated clearance pathways.14 Target‐mediated
Chiba et al
clearance primarily results from internalization of antigen–antibody immune complexes, which accelerates
clearance in a saturable manner with non‐linear pharmacokinetics.9 However, this may not be observed in healthy
volunteers because the expression or activation of the
target may be lower in healthy volunteers than in patients.
Consequently, the pharmacokinetic characteristics of
efficacy‐ and toxicity‐related antigen–antibody interactions cannot be evaluated in healthy volunteers.
Unlike mAbs, small molecule drugs are predominantly
cleared via hepatic metabolism with biliary and renal
excretion. Small molecule drugs are also capable of
penetrating tissues more effectively than mAbs, which
reflects physiological factors and the physicochemical
properties of such drugs. However, the broad distribution
of small molecule drugs contributes to their adverse
reactions in non‐target tissues (Figure 1). Thus, ethnic
differences in metabolic and physiological factors may
result in ethnic differences in the pharmacokinetics and
safety of small molecule drugs.15,16 However, this is not
necessarily true for mAbs.
From a pharmacokinetic perspective, Phase I studies in
healthy volunteers in a new region might be meaningless
for mAbs because of the absence of ethnic differences in
their distribution and elimination, similar to that of
endogenous IgG. These issues have therefore raised the
question of whether or not clinical pharmacokinetic
bridging studies are necessary for mAbs. Zhou et al5
compared the approved doses of 12 mAbs that were
approved in both Japan and the US, and discussed the
pharmacokinetic properties of mAbs in general. They
reached the conclusion that, although there is a marked
difference in body size between the two populations, this
did not necessitate different dosing regimens in the two
countries. They also highlighted the potential safety and
ethical implications of clinically unnecessary administration of mAbs to healthy subjects, a major concern because
of the large number of subjects that would be needed in
such studies. They finally recommended that it “may be
more prudent to first evaluate the available data sets
looking for potential ethnic differences in the pharmacokinetic characteristics before considering the need to
formerly conduct a clinical pharmacokinetic study.”
Phase I Studies of Monoclonal
Antibodies in Japan
The PMDA answered a question regarding the necessity of
Japanese Phase I studies before global clinical trials in a
document published by the MHLW.3 The question was “Is
it mandatory to have a Phase I trial or pharmacokinetic
information in Japanese population prior to conduct of a
global clinical trial for patients?” The PMDA’s answer to
this question was: “The dosage regimen to be used in the
global trial should be confirmed beforehand to whether it
485
does not have any particular safety problem for the
Japanese. For this purpose, before the start of the global
clinical trial, at least it is required to examine single‐dose
safety and pharmacokinetics of investigational drugs in
Japanese healthy volunteers or patients, compare the
results with non‐Japanese, and confirm that risks in the
Japanese are equivalent with non‐Japanese.” However,
they also acknowledged the possibility of a waiver for
conducting Japanese safety and pharmacokinetic studies
“If safety in the Japanese can be determined with
appropriate reasons, a Phase I trial in the Japanese is
not necessarily required prior to the global trial.”
To investigate whether or not Japanese Phase I studies
had been conducted in Japan in addition to non‐Japanese
Phase I studies, we searched the PMDA database (http://
www.info.pmda.go.jp/info/syounin_index.html)
for
mAbs that had been approved in Japan, and recorded
the study population (healthy volunteers or patients) and
the sampling type (intensive or sparse sampling).
The 24 mAbs (including mAb‐like therapeutic proteins) that had been approved in Japan as of December 2012 are listed in Table 1. The pharmacokinetic
properties in Japanese subjects were determined using
intensive blood sampling in Japanese Phase I or Phase I/II
studies for all mAbs, except for eculizumab, which was
approved for an orphan indication with limited study data
in Japanese subjects because of an extremely small patient
population in Japan (approximately 400 patients in 1998).
Most of the mAbs were approved for use in oncology
(9/24, Table 1) or to treat inflammatory diseases,
particularly rheumatoid arthritis (6/24). Phase I studies
were conducted in healthy volunteers for 1/9 mAbs used to
treat cancer and for 4/6 mAbs to treat rheumatoid arthritis.
Thus, the subject types were probably chosen according to
the target disease. Additionally, the same subject type had
been selected in non‐Japanese studies for almost all mAbs
used to treat cancer and rheumatoid arthritis. In other
words, if a study of non‐Japanese healthy volunteers had
not been conducted for a mAb, then one was not done in
Japanese healthy volunteers.
Almost all of the products were developed with either
one of the following three strategies; bridging strategies,
application of results in foreign clinical studies or using
results from global studies including Japanese patients.
Only tocilizumab and mogamulizumab underwent a full
clinical development strategy in Japan as the NDAs for
these mAbs were filed in Japan before other countries.
There were no relationships between the clinical development strategies and subjects (i.e., inclusion of healthy
volunteers or patients).
Thus, according to current guidelines, almost all of the
mAbs approved in Japan were evaluated in Japanese
pharmacokinetic studies, even though pharmacokinetic
data were available in non‐Japanese subjects. Moreover,
for 10/24 mAbs, a Phase I study was performed in
The Journal of Clinical Pharmacology / Vol 54 No 5 (2014)
486
Table 1. Therapeutic Target, Filing Strategy, and Type of Pharmacokinetic Studies Used to Support the New Drug Applications of Monoclonal
Antibodies in Japan, as of December 2012
mAb
Abatacept
Adalimumab
Basiliximab
Therapeutic target
Filing strategya
2010
2008
2010
2010
2010
2011
2012
2002
Rheumatoid arthritis
Rheumatoid arthritis
Plaque psoriasis, psoriatic arthritis
Ankylosing spondylitis
Crohn’s disease
Polyarticular juvenile idiopathic arthritis
Rheumatoid arthritis (first line)
Prophylaxis of acute organ rejection in adult patients
receiving renal transplantation
Prophylaxis of acute organ rejection in pediatric patients
receiving renal transplantation
Advanced or recurrent colorectal cancer
Advanced or recurrent colorectal cancer (additional dosing
regimen)
Advanced or recurrent non‐squamous non‐small cell lung
cancer
Breast cancer
Cryopyrin‐associated periodic syndrome
Rheumatoid arthritis (for patients with an inadequate
response to conventional therapy)
EGFR‐expressing advanced or recurrent colorectal cancer
Bone metastases from solid tumors and multiple myeloma
Paroxysmal nocturnal hemoglobinuria
Rheumatoid arthritis (for patients who have an inadequate
response to conventional therapy)
Polyarticular juvenile idiopathic arthritis
Rheumatoid arthritis (the prevention of structural joint
damage)
CD33 positive acute myeloid leukemia
Rheumatoid arthritis
Low‐grade B‐cell non‐Hodgkin’s lymphoma, mantle cell
lymphoma
Crohn’s disease
Rheumatoid arthritis
Intractable uveoretinitis
Crohn’s disease (remission maintenance)
Plaque asoriasis, psoriatic arthritis, pustular psoriasis,
erythrodermic psoriasis
Ankylosing spondylitis
Ulcerative colitis
CCR4‐positive adult T‐cell leukemia‐lymphoma
Bronchial asthma
Prevention of serious lower respiratory tract disease caused
by RSV in children
KRAS mutation‐positive colorectal cancer
Neovascular (wet) age‐related macular degeneration
Low‐grade or follicular, CD20‐positive, B‐cell non‐Hodgkin’s
lymphoma, mantle cell lymphoma (375 mg/m2 weekly for
4 doses)
CD20‐positive B‐cell non‐Hodgkin’s lymphoma new dosing
regimen (375 mg/m2 weekly for 8 doses)
Infusion prior to the administration of ibritumomab tiuxetan
Chronic idiopathic thrombocytopenic purpura
Castleman disease
Rheumatoid arthritis
Bridging study
Bridging study
Domestic study
Intensive
Intensive (HVe)
Bridging study
Intensive
2008
Bevacizumab
2007
2009
2009
Canakinumab
Certolizumab pegol
2011
2011
2012
Cetuximab
Denosumab
Eculizumab
Etanercept
2008
2012
2010
2005
2009
2012
Gemtuzumab ozogamicin
Golimumab
Ibritumomab tiuxetan
2005
2011
2008
Infliximab
2002
2003
2007
2007
2010
Mogamulizumab
Omalizumab
Palivizumab
2010
2010
2012
2009
2002
Panitumumab
Ranibizumab
Rituximab
2010
2009
2001
2003
Romiplostim
Tocilizumab
Pharmacokinetic
blood sampling
Approved
2008
2011
2005
2008
Bridging study
Bridging study
Bridging study
Intensive
Bridging study
Bridging study
Domestic study
Domestic study
Intensive (HV)
Intensive (HV)
Bridging study
Global study
Bridging study
Bridging study
Intensive
Intensive (HV)
Sparsef
Intensive (HV)
Bridging study
Domestic study
Bridging study
Domestic study
Bridging study
Intensive
Intensive (HV)
Intensive
Bridging study
Bridging study
Domestic study
Intensive
Domestic studyb
Domestic study
Bridging study
Intensive
Intensive (HV)
Intensive (HV)
Global study
Bridging study
—c
Intensive
Intensive
Intensive
Domestic study
Domestic study
Domestic study
Domestic studyb
Intensive (HV)
Intensive (HV)
(Continued)
Chiba et al
487
Table 1. Continued
mAb
Trastuzumab
Approved
Therapeutic target
2008
Polyarticular juvenile idiopathic arthritis, systemic juvenile
idiopathic arthritis
Metastatic breast cancer whose tumors overexpress the
HER2 protein (4mg/kg initially, followed by 2 mg every
week)
Breast cancer whose tumors overexpress the HER2 protein
(adjuvant)
Breast cancer whose tumors overexpress the HER2 protein
(neoadjuvant)
New dosing regimen for metastatic breast cancer whose
tumors overexpress the HER2 protein (8 mg/kg initially,
followed by 6 mg every 3 weeks)
Gastric cancer whose tumors overexpress the HER2
protein
Psoriatic arthritis, plaque psoriasis
2001
2008
2011
2011
Ustekinumab
2011
Filing strategya
Bridging study
Pharmacokinetic
blood sampling
Intensive
Global study
Special approvald
Global study
Domestic study
Intensive
CCR4, chemokine CC motif receptor 4; EGFR, endothelial growth factor receptor; HER2, human epidermal growth factor receptor 2; HV, healthy volunteers;
RSV, respiratory syncytial virus.
Including supplemental new drug applications.
a
Underlined “bridging” indicates no Japanese Phase III studies without description of extrapolation or bridging in the review report and non‐Japanese Phase III data
are referred to as “essential data”; non‐underlined “bridging” represents mAbs for which review reports include “bridging” or “extrapolation.”
b
An independent Phase I study was conducted as a first‐in‐human study.
c
Unknown whether foreign data were essential or reference.
d
Public knowledge‐based application.
e
A Phase I study in healthy Japanese volunteers was discontinued at the lowest dosage because of the emergence of adverse events (infusion site reactions).
f
Orphan drug (only 400 patients in Japan).
Japanese healthy volunteers even though data from Phase I
studies in non‐Japanese subjects were available. From an
ethical point of view, some guidelines might be necessary
to help determine whether Japanese studies in healthy
volunteers or patients with intensive sampling are
necessary before Japanese patients are enrolled in global
studies, if data from Phase I studies in non‐Japanese
subjects are available.
Evaluation of the Ethnic Differences of
Pharmacokinetics in Japanese Subjects
For some mAbs that show non‐linear PK due to target‐
mediated drug disposition, their clearance depends on the
relative expression of the target antigen. Therefore, PK
studies in healthy volunteers may not be appropriate.
Exposure information was available for 7/10 mAbs,
allowing comparison of the exposure between Japanese
and non‐Japanese healthy volunteers. Figure 2 shows the
differences in exposure and the PMDA’s assessment of the
similarity of the mAbs in healthy volunteers and patients.
Exposure (i.e., maximum plasma concentration [Cmax] and
area under the concentration–time curve [AUC]) ratios
(Japanese/non‐Japanese) in healthy volunteers ranged
from 0.6 to 1.2, which might be numerically similar.
Regarding studies in patients, data were available to
compare the exposure between Japanese and non‐
Japanese subjects for 11/14 mAbs. The PMDA concluded
that the pharmacokinetic profiles were similar for 8/11
mAbs. We calculated the Cmax and AUC (Figure 2A and
B) ratios (Japanese/non‐Japanese subjects) for these 11
mAbs, and found that the ranges of the Cmax and AUC
ratios for PMDA‐assessed “similarity” were 0.6–1.4 and
0.6–1.3, respectively. For cetuximab, although ethnic
similarity of Cmax was suspected by the regulator for the
400 mg dose (1.35‐fold), the regulator carefully commented on the similarity in the expanded indication
for head and neck cancer. Therefore, a 1.35‐fold difference might represent the regulator’s upper limit for
assuming similarity. Only three mAbs had values beyond
these ranges: ustekinumab (90 mg) (Cmax ratio: 1.56),
rituximab (Cmax: 0.38, AUC: 0.42) and ibritumomab
tiuxetan (AUC: 1.84). However, there were some
exceptions, even within the range (basiliximab [20 mg]
and ustekinumab [45 mg]).
Table 2 lists the mAbs that the PMDA judged to be
non‐similar in healthy volunteers and patients, along with
the reason written in the common technical document
(CTD) or PMDA review reports why the sponsors and the
PMDA considered the pharmacokinetics to be “non‐
similar” (details in Supplemental Table S1).
Despite the “non‐similar” evaluations of omalizumab,
basiliximab (20 mg), ibritumomab tiuxetan, rituximab,
and ustekinumab, their doses used in the subsequent
The Journal of Clinical Pharmacology / Vol 54 No 5 (2014)
488
0.4
0.6
0.8
1
1.2
1.4
1.6
1.8
2
0.2
Palivizumab (3 mg/kg)
Certolizumab pegol (400 mg)
Etanercept (50 mg)
Etanercept (25 mg)
Denosumab (3 mg/kg)
Etanercept (10 mg)
Omalizumab (150 mg)
Golimumab (100 mg)
Denosumab (1 mg/kg)
Rituximab (375 mg/m2)
Abatacept (2 mg/kg)
Infliximab (5 mg/kg, 10 mg/kg)
Bevacizumab (10 mg/kg)
Bevacizumab (15 mg/kg)
Adalimumab (80 mg)
Panitumumab (6 mg/kg)
Abatacept (10 mg/kg)
Adalimumab (40 mg)
Ustekinumab (90 mg)
Ustekinumab (45 mg)
Cetuximab (250 mg/m2)
Gemtuzumab ozogamicin (9 mg/m2)
Cetuximab (400 mg/m2)
Ibritumomab tiuxetan (14.8 MBq/Kg)
Ibritumomab tiuxetan (11.1 MBq/Kg)
Rituximab (375 mg/m2)
Abatacept (2 mg/kg)
0.6
0.8
1
1.2
1.4
1.6
䕕Similar
䕔Non-similar
Canakinumab (150 mg)
Golimumab (50 mg)
Adalimumab (80 mg)
Panitumumab (6 mg/kg)
Abatacept (10 mg/kg)
Adalimumab (40 mg)
Ustekinumab (90 mg)
Ustekinumab (45 mg)
Cetuximab (250 mg/m2)
Gemtuzumab ozogamicin (9 mg/m2)
Cetuximab (400 mg/m2)
Patients
Healthy volunteers
Palivizumab (3 mg/kg)
Certolizumab pegol (400 mg)
Etanercept (50 mg)
Etanercept (25 mg)
Denosumab (3 mg/kg)
Etanercept (10 mg)
Omalizumab (150 mg)
Golimumab (100 mg)
Denosumab (1 mg/kg)
Palivizumab (15 mg/kg)
Canakinumab (150 mg)
Golimumab (50 mg)
0.4
Basiliximab (20 mg)
0.4
0.6
0.8
1
1.2
1.4
1.6
1.8
2
AUC ratio
0.2
0.4
0.6
0.8
1
1.2
1.4
1.6
Cmax ratio
Figure 2. Area under the concentration–time curve (AUC) ratios (left panel) and maximum plasma concentration (Cmax) ratios (right panel) for
monoclonal antibodies (mAbs) between Japanese and non‐Japanese subjects. Upper and lower figures expressed for healthy volunteers and patients,
respectively. Underlined mAbs target cell surface antigens; mAbs without underlining target soluble antigens. The data were extracted from the studies
in which the pharmacokinetic properties were compared between Japanese and non‐Japanese subjects as of December 2012. If multiple mean values
were reported for the same dose, the overall weighted geometric mean was calculated. If the PMDA clearly concluded that there was an ethnic difference
in the pharmacokinetics profiles or if the PMDA agreed or did not deny an apparent/statistically significant ethnic difference in pharmacokinetic
properties reported in the common technical documents (CTDs), the pharmacokinetics characteristics were reported as “non‐similar” (black columns),
otherwise they were reported as “similar” (white columns). The dashed lines represent the upper and lower bounds of similarity. Eculizumab is not listed
because the Cmax and AUC values were not reported for pharmacokinetic comparisons in the review report or CTD but the ratio of CL was 1.17.
Ranibizumab, trastuzumab, tocilizumab, romiplostim, and mogamulizumab are not listed because no pharmacokinetic data were available for the
comparison or the pharmacokinetics were non‐linear.
clinical studies in Japan were the same as those stated on
the approved US labels, and the dose regimens stated on
the US and Japan labels are essentially similar (Table 2).
The regulatory assessment for omalizumab, basiliximab, and usutekinumab was “non‐similar,” even though
there were small numerical differences in pharmacokinetic
parameters (Figure 2). For omalizumab, we could not find
any pharmacokinetic factors that could account for the
“non‐similar” evaluation. For basiliximab, there were no
ethnic differences in Cmax, while differences in Vss and
CL were explained by body weight. For ustekinumab
(45 mg), the regulatory assessment was possibly influenced by the results reported for ustekinumab (90 mg), for
which there was an apparent ethnic difference in Cmax.
For the three mAbs that had values beyond the ranges
of “similarity” (Cmax 0.6–1.4, AUC 0.6–1.3), ibritumomab tiuxetan and rituximab both target cell‐surface
antigens, which influences mAb clearance. By contrast,
ustekinumab targets a soluble antigen, which hardly
affects mAb clearance. The ethnic differences in the
pharmacokinetics of ibritumomab tiuxetan and rituximab
were explained by CD20 levels. Ethnic differences in cell
surface expression of the molecular targets of rituximab
and ibritumomab tiuxetan could affect their exposure
because the pharmacokinetics of mAbs are associated with
the immune complex concentration. For ustekinumab, the
NDA review report described that the difference in its
pharmacokinetics could be explained by body weight. In
fact, the Cmax, which was affected by distribution volume,
showed ethnic differences whereas AUC did not.
In summary, the observed differences in pharmacokinetics between Japanese and non‐Japanese were either due
to differences in body weight or differences in receptor
expression between the populations. The body weight
difference between Japanese and Caucasian individuals is
only about 20%,16 but the difference in receptor
expression level depends on the target disease. If the
difference is covered by the safety margin, dose
adjustment might not be necessary, as was observed for
clinical dose shown in Table 2.
Safety of Monoclonal Antibodies in
Healthy Volunteers
As the pharmacokinetics of mAbs are similar between
Japanese and non‐Japanese subjects, and because the
target is usually less detectable in healthy volunteers than
in patients, the safety profiles of mAbs in healthy
volunteers should also be similar between Japanese and
non‐Japanese subjects. Table 3 summarizes the adverse
45 mg initially and 4 weeks
later, followed by
45 mg every 12 weeks
(body weight 100 kg)
90 mg initially and 4 weeks
later, followed by
90 mg every 12 weeks
(body weight >100 kg)
150–375 mg every 2 or
4 weeks (adjustment by
IgE and body weight)
First 20 mg within 2 h
before transplantation,
followed by 20 mg at
4 days after
transplantation
14.8 MBq/kg for patients
with normal platelet
count
11.1 MBq/kg for patients
with platelet count of
100,000–149,000 cells/
mm3
375 mg/m2 weekly for 4
or 8 doses
Non‐Japanese
20 mg
14.8 MBq/kg
11.1 MBq/kg
375 mg/m2
20 mg (patients)
14.8 MBq/kg (patients)
11.1 MBq/kg (patients)
375 mg/m2 (patients)
45 mg (patients)
90 mg (patients)
No difference
No difference
45 mg initially and 4 weeks
later, followed by
45 mg every 12 weeks
For patients who have an
incomplete response,
90 mg every 12 weeks
(similar regimen)g
90 mg
45 mg
150–375 mg every 2 or 4
weeks
Subsequent phases in
patientse
150 mg (healthy
volunteers)
Pharmacokinetic study
phasesd
75–375 mg every 2 or
4 weeks (adjustment by
IgE and body weight)f
No difference
Japanese
Dose
1.84
—h
1.56
1.2
1.05
1.12
0.42
1.48
—h
0.38
—h
0.94
AUC
1.09
1.17
Cmax
Ratiob
Sponsor
Non‐similar: it is probably
caused by differences in
CD20 expression
Non‐similar: caused by
differences in body
weight
Non‐similar: caused by
differences in CD20
expression
Non‐similar: the reason
proposed by the
sponsor was accepted
Non‐similar: the cause of
the difference was not
determined
Non‐similar: the cause of
the difference was not
determined
Non‐similar: the cause of
the difference was not
determined
Non‐similar: it is difficult
to confirm similarity
Regulator
Assessment of similarityc
Non‐similar: caused by IgE
concentration
difference
Non‐similar: caused by
differences in body
weight
AUC, area under the concentration–time curve; Cmax, maximum plasma concentration; mAb, monoclonal antibody.
The mAbs included in this table are evaluated as “non‐similar” in PMDA review reports as of December 2012 (see legends in Figure 2).
a
Dose regimens stated on the US and Japanese labels.
b
Cmax ratios and AUC ratios between Japanese and non‐Japanese subjects described in Figure 2.
c
The assessments of similarity are described based on the CTD (sponsor evaluation) and the PMDA review report (PMDA evaluation).
d
Dose in the pharmacokinetic studies used to compare pharmacokinetic properties.
e
Doses used in subsequent clinical studies, which were conducted after the pharmacokinetic studies.
f
Dose adjustments based on the patient’s bodyweight and IgE concentration were recommended by the PMDA.
g
Body weight‐tiered dose regimens were not recommended by the PMDA because of the small number of patients with a body weight >100 kg in Japanese clinical trials.
h
Not reported.
Ustekinumab
Rituximab
Ibritumomab
tiuxetan
Basiliximab
Omalizumab
mAb
Recommended dosea
Table 2. Relationship Between the Dose Used in Pharmacokinetic Studies and Doses Used in Subsequent Clinical Studies for mAbs Evaluated as being “Non‐Similar,” as of December 2012
Chiba et al
489
The Journal of Clinical Pharmacology / Vol 54 No 5 (2014)
490
Table 3. Summary of Adverse Reactions Associated With Monoclonal Antibodies in Healthy Japanese Volunteers, as of December 2012
mAb
Adalimumab
Canakinumab
Dose
0.1 mg/kg
1 mg/kg i.v.
3 mg/kg i.v.
600 mg i.v.
150 mg s.c.
300 mg s.c.
600 mg i.v. þ 300 mg s.c.
Placebo
Certolizumab pegol
100 mg
400 mg
800 mg
Placebo
Denosmab
0.03 mg/kg
0.1 mg/kg
0.3 mg/kg
1.0 mg/kg
3.0 mg/kg
Placebo
Etanercept
10 mg
25 mg
Golimumab
50 mg
placebo
50 mg
100 mg
All adverse event (number of patients)
Rash (7/15), leukopenia (1/15)
Laboratory test abnormal (2/6), injury, poisoning and procedural
complications (1/6)
Gastrointestinal disorders (2/6), infections and infestations (1/6)
None
Gastrointestinal disorders (2/6), infections and infestations (1/6)
Laboratory test abnormal (4/6)
Laboratory test abnormal (2/6), eye disorders (1/6)
Laboratory test abnormal (7/12), infections and infestations (2/12),
gastrointestinal disorders (2/12)
Queasy (1/6), nasopharyngitis (1/6)
Nasopharyngitis (1/6), chickenpox (2/6), headache NOS (2/6)
Queasy (2/6), sore throat NOS (2/6), influenza like illness (1/6),
nasopharyngitis (2/6), back pain (2/6), headache NOS (3/6)
Sore throat NOS (1/6), Injection site pain (2/6), nasopharyngitis (1/6),
headache NOS (2/6)
Nasopharyngitis (1/6), chest discomfort (1/6)
Nasopharyngitis (2/6), back pain (1/6), atrial fibrillation (1/6),
dizziness (1/6), gastritis (1/6)
Rhinorrhea (1/6), abdominal pain (1/6), arthralgia (1/6), chest
discomfort (1/6), headache (1/6), malaise (1/6), stomatitis (1/6),
cheilitis (1/6), conjunctival hemorrhage (1/6), contusion (1/6),
dermatitis contact (1/6), herpes zoster (1/6), hypoesthesia (1/6),
seasonal allergy (1/6), sensation of block in ear (1/6), tinnitus (1/6)
Nasopharyngitis (5/6), rhinorrhea (2/6), injection site pain (1/6),
abdominal pain (1/6), arthralgia (1/6), back pain (1/6), eczema
(1/6), headache (1/6), malaise (1/6), stomatitis (1/6), thermal burn
(1/6), bronchitis acute (1/6), diarrhea (1/6), fatigue (1/6), muscle
fatigue (1/6), rash (1/6)
Injection site pain (2/6), nasopharyngitis (1/6), eczema (1/6), thermal
burn (1/6), blepharospasm (1/6), blister (1/6), conjunctivitis (1/6),
hemorrhage subcutaneous (1/6), injection site erythema (1/6),
periarthritis (1/6), periodontal infection (1/6), spondylosis (1/6)
Arthralgia (1/10), contusion (1/10), muscle fatigue (1/10), tinnitus
(1/10), chest pain (1/10), dermatitis (1/10), gingival bleeding (1/10),
hepatic enzyme increased (1/10), injection site hemorrhage (1/10),
lip blister (1/10), myalgia (1/10), pruritus (1/10), rhinitis allergic
(1/10), stomach discomfort (1/10)
Nasal discharge (2/8), pain pharynx (1/8), general malaise (1/8),
twilight state (1/8), CRP increased (1/8), cough (1/8), pharynx
redness (1/8), chills (1/8), dull headache (1/8), Abdominal
distension (1/8), headaches (1/8), cough (1/8), white blood cell
decreased (1/8), white blood cell increased (1/8)
Nasal discharge (2/8), triglyceride increase (2/8), pain pharynx (1/8),
general malaise (1/8), twilight state (1/8), CRP increased (1/8),
fever (1/8), hot feeling generalized (1/8), wooziness), (1/8),
paroxysmal sneeze (1/8), tachycardia (1/8), hot flush (1/8),
monocyte count increased (1/8), lymphocyte count decreased
(1/8)
Triglyceride increase (3/8), nasal discharge (1/8), low back ache (1/8)
Pain pharynx (1/6), cough (1/6)
Gastrointestinal disorders respiratory (2/12), thoracic and
mediastinal disorders (2/12), infections and infestations (2/12),
injury, poisoning and procedural complications (1/12), nervous
system disorders (1/12)
Gastrointestinal disorders respiratory (4/12), thoracic and
mediastinal disorders (2/12), infections and infestations (2/12)
Severity of adverse event
Slight or moderate
Slight or moderate
No severe AEa
Slight or moderate
Slight
(Continued)
Chiba et al
491
Table 3. Continued
mAb
Dose
Omalizumab
150 mg
Omalizumab
75 mg
150 mg
300 mg
375 mg
Placebo
Palivizumab
3 mg/kg
10 mg/kg
15 mg/kg
Romiplostim
0.3 mg/kg
1 mg/kg
2 mg/kg
Placebo
Tocilizumab
0.15 mg/kg
0.5 mg/kg
1 mg/kg
All adverse event (number of patients)
Laboratory test abnormal (1/20), skin and subcutaneous tissue
disorders (1/20)
Respiratory, thoracic and mediastinal disorders (5/12), general
disorders and administration site conditions (2/12),
gastrointestinal disorders (1/12), skin and subcutaneous tissue
disorders (1/12)
Respiratory, thoracic and mediastinal disorders (1/12), skin and
subcutaneous tissue disorders (1/12)
Respiratory, thoracic and mediastinal disorders (4/12),
gastrointestinal disorders (2/12), general disorders and
administration site conditions (1/12), nervous system disorders (1/
12)
Respiratory, thoracic and mediastinal disorders (5/12), general
disorders and administration site conditions (1/12),
gastrointestinal disorders (1/12), nervous system disorders (1/12),
skin and subcutaneous tissue disorders (1/12), musculoskeletal
and connective tissue and bone disorders (1/12)
Respiratory, thoracic and mediastinal disorders (7/24), general
disorders and administration site conditions (4/24), nervous
system disorders (3/24), musculoskeletal and connective tissue
and bone disorders (2/24), gastrointestinal disorders (2/24),
cardiac disorders (1/24), infections and infestations (1/24), renal
and urinary disorders (1/24), skin and subcutaneous tissue
disorders (1/24)
Respiratory system disorder (4/12), urogenital diseases (3/12),
general disorders and administration site conditions (2/12),
enterogastric disorder (1/12), blood and lymphatic system
disorders (1/12)
Urogenital diseases (1/6)
Respiratory system disorder (3/6), general disorders and
administration site conditions (2/6), enterogastric disorder (2/6)
Laboratory test abnormal (4/6), gastrointestinal disorders (1/6),
general disorders and administration site conditions (1/6),
musculoskeletal and connective tissue disorders (1/6), nervous
system disorders (1/6)
Gastrointestinal disorders (3/6), general disorders and administration
site conditions (2/6), nervous system disorders (2/6), laboratory
test abnormal (1/6), metabolism and nutrition disorders (1/6),
musculoskeletal and connective tissue disorders (1/6)
Infections and infestations (3/6), gastrointestinal
disorders (2/6), laboratory test abnormal (2/6), musculoskeletal
and connective tissue disorders (2/6), nervous system
disorders (1/6)
Gastrointestinal disorders (2/6), general disorders and administration
site conditions (1/6), immune system disorders (1/6), infections
and infestations (1/6), laboratory test abnormal (1/6)
Eye disorders (1/5), gastrointestinal disorders (1/5), laboratory test
abnormal (1/5)
Laboratory test abnormal (3/5), gastrointestinal disorders (2/5),
infections and infestations (1/5), eye disorders (1/5), respiratory,
thoracic and mediastinal disorders (1/5), musculoskeletal and
connective tissue disorders (1/5), general disorders and
administration site conditions (1/5)
Respiratory, thoracic and mediastinal disorders (3/5), gastrointestinal
disorders (3/5), musculoskeletal and connective tissue disorders
(3/5), laboratory test abnormal (3/5), general disorders and
administration site conditions (2/5)
Severity of adverse event
No severe AEb
Slight or moderate
Slight or moderate
Slight or moderate
Slight
Moderate
Moderate
(Continued)
The Journal of Clinical Pharmacology / Vol 54 No 5 (2014)
492
Table 3. Continued
mAb
Dose
2 mg/kg
Placebo
All adverse event (number of patients)
Immune system disorders (4/5), general disorders and administration
site conditions (2/5), laboratory test abnormal (2/5), respiratory,
thoracic, and mediastinal disorders (1/5)
Immune system disorders (3/8), respiratory, thoracic, and
mediastinal disorders (3/8), gastrointestinal disorders (3/8),
general disorders and administration site conditions (2/8),
psychiatric disorders (1/8), eye disorders (1/8), musculoskeletal
and connective tissue disorders (1/8), laboratory test abnormal (1/
8), injury, poisoning, and procedural complications (1/8)
Severity of adverse event
Slight
Slight
AE, adverse event; CRP, C‐reactive protein; i.v., intravenous; mAb, monoclonal antibody; NOS, not otherwise stated; s.c., subcutaneous.
The common technical document (CTD) is unavailable.
b
There is no description about severity in the CTD.
a
reactions observed in Phase I studies involving healthy
Japanese volunteers. As indicated in this table, the
majority of adverse reactions were classified as slight,
with only a few moderate events. No severe adverse
reactions were reported in any of the studies.
Discussion
The PMDA has accumulated much data through evaluation of drugs developed by bridging strategies that can
allow for comparisons of ethnic differences or similarities,
and may allow us to predict the impact of ethnicity for new
compounds.
In this review, we first examined the study type and
sampling methods used in the Japanese clinical pharmacokinetic studies of mAbs, as reported in the PMDA’s
publically available database of NDAs. The sampling
methods of all mAbs, except for eculizumab, involved
intensive blood sampling, suggesting that the studies were
classified as pharmacokinetic/safety studies. Although the
“Basic Principles on Global Clinical Trials” acknowledges the possibility of a waiver for pharmacokinetic/
safety studies, Japanese pharmacokinetic/safety studies
are still required before Japanese patients can be enrolled
in global clinical trials.
Simultaneous filing of a NDA with global clinical trials
is becoming increasingly common in Japan and Asia.
Drug development programs generally start in the US/EU,
and Asian populations are ideally incorporated after
establishing the proof of concept to reduce the risk of
attrition. However, if regional pharmacokinetic/safety
studies are essential, it may be difficult for the sponsor to
include time for such studies between the first‐in‐human
and proof of concept studies. Additionally, from ethical
and efficiency perspectives, it might be difficult to expect
clinically meaningful results from Japanese pharmacokinetic/safety studies for mAbs in particular, because the
pharmacokinetics of mAbs are dependent on the target
levels, which could be measured in diagnostic practice.
In the present review, we retrieved the PMDA’s
evaluation of each mAb from the CTD and review reports.
Through this review, we found that the scientific
discussion regarding the apparent ethnic differences for
omalizumab, ibritumomab tiuxetan, and rituximab focused on the possible differences in antigen levels between
Japanese and non‐Japanese subjects. Of course, it is
difficult to exclude possible ethnic differences in genetic
factors, and differences in antigen levels might contribute
to the different pharmacokinetic/pharmacodynamic results. We also found that the pharmacokinetics of mAbs
targeting soluble antigens were similar between Japanese
and non‐Japanese subjects, except for omalizumab and
ustekinumab, which were comparable either numerically
or after considering body weight. The effects of body size
on the pharmacokinetics of mAbs were discussed in the
context of adjusting the doses of mAbs, and it was
suggested that body weight had little or only moderate
effects on the doses of most mAbs.17,18
Of note, studies in healthy Japanese volunteers may not
provide any additional insight into the pharmacokinetic
properties of a mAb beyond that obtained in prior Phase I
studies of non‐Japanese subjects. Actually, the doses
selected in subsequent Japanese patient phases were the
same as those stated on the approved US labels, even if
there were differences in pharmacokinetics in the healthy
volunteer studies (omalizumab, Table 2). In this review,
we confirmed that there are essentially no differences in
exposure between Japanese and non‐Japanese subjects,
and that there are no safety concerns specific to healthy
Japanese volunteers for the mAbs approved in Japan.
Keizer et al8 suggested that Phase I studies in healthy
volunteers may provide only limited information regarding the pharmacokinetics characteristics of mAbs.
Additionally, the need for fractionated doses of many
mAbs means that the pharmacokinetic characteristics
determined in Phase I studies may not truly represent the
pharmacokinetics in patients. Although the incidence of
severe adverse reactions may be low for many mAbs,
Chiba et al
493
mAb
Are PK
characteristics linear?
Yes
No
Considering the safety, is an
ethnic difference of the
antigen level predictable?
Yes
Predict the antigen level and
pharmacokinetics
Is there any exposureindependent toxicity?b
No
Safety (PK) study in patientsa
(intensive sampling)
No
yes
Skip Japanese phase I studies
Safety studyc
Sparse PK sampling can be
performed in Phase II studies
Figure 3. Proposed algorithm for assessing whether Phase I studies in
Japanese patients could be waived for monoclonal antibodies if data from
prior non‐Japanese studies are available. aIntensive blood sampling may be
performed in Phase IIa studies instead of Phase I studies. bThe need for
safety studies to detect exposure‐independent toxicity in Japanese
patients should be discussed with the Pharmaceutical and Medical
Devices Agency. cConsidering Japanese‐specific antigen‐independent
serious adverse events, studies (possibly Phase II) should be conducted
with careful monitoring of safety. Studies in other Asian countries might
be useful to predict safety profiles in Japanese patients.
reducing the exposure of healthy volunteers to clinically
relevant doses of mAbs is important from a safety
perspective.
Based on these issues, we suggest that Phase I studies in
Japanese healthy volunteers could be waived. Moreover,
Phase I‐type studies in Japanese patients could be waived
for some mAbs if suitable studies have already been
performed in other countries. Figure 3 presents an
algorithm that could help determine whether or not such
studies could be waived.
First, from a pharmacokinetic perspective, if the mAb
shows linear pharmacokinetics within the anticipated
therapeutic dose range in prior non‐Japanese Phase I
studies, almost all antigens must form a complex with
excessive mAbs. Hence, low antigen levels do not affect
the pharmacokinetics of the mAbs. If the mAb shows a
non‐linear profile, its pharmacokinetic properties are most
likely affected by the target levels. In this case, if the target
level can be measured or predicted, the pharmacokinetic
properties of the mAb can be predicted in Japanese
subjects. If the target level cannot be measured or
predicted, then it is necessary to perform appropriate
pharmacokinetic/safety studies in Japanese subjects.
Next, from a safety perspective, if the pharmacokinetics of a mAb are linear or can be predicted from prior
studies in non‐Japanese subjects, then the sponsor must
consider the risk of exposure‐independent toxicity due to
off‐target toxicity. If this is the case, the toxicity will not be
predictable and safety studies might be necessary in
Japanese subjects. However, it might be necessary to
conduct such studies in the target patients considering the
benefit/risk balance. By contrast, if exposure‐independent
toxicity is not apparent, pharmacokinetic/safety studies
involving intensive sampling in healthy volunteers and
patients may be unnecessary.
In conclusion, the present analyses revealed no
essential differences between Japanese and non‐Japanese
subjects, particularly in healthy volunteers, in the
exposure of mAbs that have been approved in Japan.
The PMDA assessment of similarity was reasonable, as
the ranges of Cmax and AUC ratios were 0.6–1.4 and 0.6–
1.3, respectively, although there were a few exceptions.
Moreover, most of the observed differences in PK were
accounted for by differences in body weight or in target
expression levels between Japanese and non‐Japanese
subjects. The studies entailed intensive blood sampling,
except for one mAb in which sparse sampling methods
were used. Studies in healthy volunteers were conducted
for approximately half of the mAbs. Based on these data,
we think that a waiver for Japanese pharmacokinetic/
safety studies following non‐Japanese Phase I studies will
become more commonplace. Our review should help in
the evaluation of mAbs currently under development, and
to determine the necessity for Japanese Phase I pharmacokinetic and safety studies.
Acknowledgments
We wish to thank Shinichi Tsuchiwata, MSc, for scientific advice
and Nicholas D. Smith, PhD, for providing editorial support.
Declaration of Conflicting Interests
Hiroyuki Yoshitsugu was an employee of Bristol‐Myers Squibb
Co., Ltd. at the time of this research. Masaki Hiraoka is an
employee of Bristol‐Myers K.K. Satofumi Iida and Koichiro
Yoneyama are employees of Chugai Pharmaceutical Co., Ltd.
Takahiko Tanigawa is an employee of Bayer Pharma AG. The
authors have no stock options or other incentives in addition to
research support and consulting agreements.
Author Contributions
Koji Chiba and Hiroyuki Yoshitsugu designed
research method; all of the authors performed
analyzed the research and results; Koji Chiba
Takahiko Tanigawa wrote the manuscript; all of
authors read and reviewed the manuscript.
the
and
and
the
References
1. International Conferrence on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. ICH
Harmonised Tripartite Guideline: Ethnic Factors in the Acceptability
of Foreign Clinical Data: E5(R1) 1998.
2. Pharmaceutical and Medical Safety Bureau, Ministry of Health,
Labour and Welfare, Japan. Notification No. 739 (kyokuchou‐tsuuchi).
August 11. Tokyo: Ministry of Health, Labour and Welfare; 1998.
3. Evaluation and Licensing Division, Pharmaceutical and Food Safety
Bureau, Ministry of Health, Labour and Welfare, Japan. Basic
Principles on Global Clinical Trials. Notification no. 0928010; 2007.
494
4.
5.
6.
7.
8.
9.
10.
11.
http://www.pmda.go.jp/regulatory/file/guideline/new_drug/GlobalClinicalTrials_en.pdf.
Ichimaru K, Toyoshima S, Uyama Y. Effective global drug
development strategy for obtaining regulatory approval in Japan
in the context of ethnicity‐related drug response factors. Clin
Pharmacol Ther. 2010;87:362–366.
Zhou H, Tsukamoto Y, Davis HM. Should clinical pharmacokinetic
bridging studies between Caucasian and Asian populations be
required for approval of monoclonal antibodies? J Clin Pharmacol.
2012;52:1273–1276.
Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther. 2008;84:
548–558.
Brambell FW. The transmission of immunity from mother to young
and the catabolism of immunoglobulins. Lancet. 1966;2:1087–1093.
Keizer RJ, Huitema AD, Schellens JH, Beijnen JH. Clinical
pharmacokinetics of therapeutic monoclonal antibodies. Clin
Pharmacokinet. 2010;49:493–507.
Mould DR, Green B. Pharmacokinetics and pharmacodynamics of
monoclonal antibodies: concepts and lessons for drug development.
BioDrugs. 2010;24:23–39.
Ishii‐Watabe A, Saito Y, Suzuki T, et al. Genetic polymorphisms of
FCGRT encoding FcRn in a Japanese population and their functional
analysis. Drug Metab Pharmacokinet. 2010;25:578–587.
Smith KG, Clatworthy MR. FcgammaRIIB in autoimmunity and
infection: evolutionary and therapeutic implications. Nat Rev
Immunol. 2010;10:328–343.
The Journal of Clinical Pharmacology / Vol 54 No 5 (2014)
12. Takai T. Roles of Fc receptors in autoimmunity. Nat Rev Immunol.
2002;2:580–592.
13. Torkildsen O, Utsi E, Mellgren SI, Harbo HF, Vedeler CA, Myhr
KM. Ethnic variation of Fc gamma receptor polymorphism in Sami
and Norwegian populations. Immunology. 2005;115:416–421.
14. Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of
therapeutic monoclonal antibodies. Drug Discov Today.
2006;11:81–88.
15. Arnold FL, Kusama M, Ono S. Exploring differences in drug doses
between Japan and Western countries. Clin Pharmacol Ther.
2010;87:714–720.
16. Myrand SP, Sekiguchi K, Man MZ, et al. Pharmacokinetics/
genotype associations for major cytochrome P450 enzymes in native
and first‐ and third‐generation Japanese populations: comparison
with Korean, Chinese, and Caucasian populations. Clin Pharmacol
Ther. 2008;84:347–361.
17. Bai S, Jorga K, Xin Y, et al. A guide to rational dosing of monoclonal
antibodies. Clin Pharmacokinet. 2012;51:119–135.
18. Wang DD, Zhang S, Zhao H, Men AY, Parivar K. Fixed dosing
versus body size‐based dosing of monoclonal antibodies in adult
clinical trials. J Clin Pharmacol. 2009;49:1012–1024.
Supporting Information
Additional supporting information may be found in the
online version of this article at the publisher’s web‐site.
Download