huang 2013

12
MINI REVIEWS The Utility of Modeling and Simulation in Drug Development and Regulatory Review SHIEW-MEI HUANG, DARRELL R. ABERNETHY, YANING WANG, PING ZHAO, ISSAM ZINEH Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland Received 1 March 2013; revised 6 April 2013; accepted 9 April 2013 Published online 24 May 2013 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23570 ABSTRACT: US Food and Drug Administration (FDA) has identified innovation in clinical evaluations as a major scientific priority area. This paper provides case studies and updates to describe the efforts by the FDA’s Office of Clinical Pharmacology in its development and appli- cation of regulatory science, focusing on modeling and simulation. Key issues and challenges are identified that need to be addressed to promote the uptake of modeling and simulation approaches in drug regulation. Published 2013. This article is a U.S. Government work and is in the public domain in the USA. 102:2912–2923, 2013 Keywords: regulatory science; PBPK; Modeling and simulation; systems pharmacology; phar- macodynamics; drug interactions; dose-response; pharamcokinetics INTRODUCTION One important aspect of the United States Food and Drug Administration (US FDA)’s mission is to make drug therapies available to the American public in a timely manner. In its recent efforts to foster ef- ficient and informative drug development, the US FDA has made priorities of promoting biomedical innovation, early communication with drug develop- ers, and administrative and scientific flexibility. 1 The US FDA approved 35 new molecular entities in fis- cal year 2012. Approvals included groundbreaking treatments for a variety of unmet medical needs. (Table 1) Additionally, many of these therapies were developed, evaluated by US FDA, and/or approved via expedited mechanisms. Despite these successes, there have been intermittent calls to reform the drug de- velopment and regulatory enterprises (Ref. 2 and ref- erences therein). Provisions in the recently enacted US FDA Safety and Innovation Act (FDASIA) 3 and the reauthorized Prescription Drug User Fee Act V (PDUFA V) underscore the need for and importance of developing new approaches to streamline drug de- velopment and regulatory evaluation. Various pub- lic–private partnerships have been formed under the Correspondence to: Shiew-Mei Huang (Telephone: +301- 796-1541; Fax: +301-847-8720; E-mail: ShiewMei.Huang @FDA.HHS.GOV) Journal of Pharmaceutical Sciences, Vol. 102, 2912–2923 (2013) Published 2013. This article is a U.S. Government work and is in the public domain in the USA. “critical path initiatives” 4,5 to improve the exchange of innovations and information about drug develop- ment among the US FDA, industry, academic scien- tists, and patient advocacy groups. A recent update on these activities has shown results of leveraging collaborations in regulatory science. 6 The United States Food and Drug Administration continues to target improvements in regulatory sci- ence, including the development of scientific tools that can bridge the gap between cutting-edge discoveries and real-world diagnostics and therapeutics. US FDA has identified innovation in clinical evaluations (e.g., through modeling and simulation) as a major scien- tific priority area. 7 US FDA’s Office of Clinical Phar- macology (OCP) has used modeling and simulation strategies to address a variety of drug development, regulatory, and therapeutic questions over the past decade. 8–11 Notwithstanding, the science of quantita- tive clinical pharmacology continues to advance at a rapid pace such that regulators must constantly eval- uate the most appropriate applications of modeling, simulation, and other innovations in the public health context. 2 In this paper, we discuss recent efforts by the US FDA’s OCP in the development and application of reg- ulatory science focusing on modeling and simulation. Case studies and updates are provided to illustrate (1) the impact of pharmacometric analyses on the premarket approval and labeling of new drugs and the application of accumulated regulatory experience to the review of investigational new drugs (INDs), 2912 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Upload: gusgara

Post on 27-Jul-2016

226 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Huang 2013

MINI REVIEWS

The Utility of Modeling and Simulation in Drug Developmentand Regulatory Review

SHIEW-MEI HUANG, DARRELL R. ABERNETHY, YANING WANG, PING ZHAO, ISSAM ZINEH

Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and DrugAdministration, Silver Spring, Maryland

Received 1 March 2013; revised 6 April 2013; accepted 9 April 2013

Published online 24 May 2013 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23570

ABSTRACT: US Food and Drug Administration (FDA) has identified innovation in clinicalevaluations as a major scientific priority area. This paper provides case studies and updates todescribe the efforts by the FDA’s Office of Clinical Pharmacology in its development and appli-cation of regulatory science, focusing on modeling and simulation. Key issues and challengesare identified that need to be addressed to promote the uptake of modeling and simulationapproaches in drug regulation. Published 2013. This article is a U.S. Government work and isin the public domain in the USA. 102:2912–2923, 2013Keywords: regulatory science; PBPK; Modeling and simulation; systems pharmacology; phar-macodynamics; drug interactions; dose-response; pharamcokinetics

INTRODUCTION

One important aspect of the United States Food andDrug Administration (US FDA)’s mission is to makedrug therapies available to the American public ina timely manner. In its recent efforts to foster ef-ficient and informative drug development, the USFDA has made priorities of promoting biomedicalinnovation, early communication with drug develop-ers, and administrative and scientific flexibility.1 TheUS FDA approved 35 new molecular entities in fis-cal year 2012. Approvals included groundbreakingtreatments for a variety of unmet medical needs.(Table 1) Additionally, many of these therapies weredeveloped, evaluated by US FDA, and/or approved viaexpedited mechanisms. Despite these successes, therehave been intermittent calls to reform the drug de-velopment and regulatory enterprises (Ref. 2 and ref-erences therein). Provisions in the recently enactedUS FDA Safety and Innovation Act (FDASIA)3 andthe reauthorized Prescription Drug User Fee Act V(PDUFA V) underscore the need for and importanceof developing new approaches to streamline drug de-velopment and regulatory evaluation. Various pub-lic–private partnerships have been formed under the

Correspondence to: Shiew-Mei Huang (Telephone: +301-796-1541; Fax: +301-847-8720; E-mail: [email protected])Journal of Pharmaceutical Sciences, Vol. 102, 2912–2923 (2013)Published 2013. This article is a U.S. Government work and is in the publicdomain in the USA.

“critical path initiatives”4,5 to improve the exchangeof innovations and information about drug develop-ment among the US FDA, industry, academic scien-tists, and patient advocacy groups. A recent updateon these activities has shown results of leveragingcollaborations in regulatory science.6

The United States Food and Drug Administrationcontinues to target improvements in regulatory sci-ence, including the development of scientific tools thatcan bridge the gap between cutting-edge discoveriesand real-world diagnostics and therapeutics. US FDAhas identified innovation in clinical evaluations (e.g.,through modeling and simulation) as a major scien-tific priority area.7 US FDA’s Office of Clinical Phar-macology (OCP) has used modeling and simulationstrategies to address a variety of drug development,regulatory, and therapeutic questions over the pastdecade.8–11 Notwithstanding, the science of quantita-tive clinical pharmacology continues to advance at arapid pace such that regulators must constantly eval-uate the most appropriate applications of modeling,simulation, and other innovations in the public healthcontext.2

In this paper, we discuss recent efforts by the USFDA’s OCP in the development and application of reg-ulatory science focusing on modeling and simulation.Case studies and updates are provided to illustrate(1) the impact of pharmacometric analyses on thepremarket approval and labeling of new drugs andthe application of accumulated regulatory experienceto the review of investigational new drugs (INDs),

2912 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Page 2: Huang 2013

THE UTILITY OF MODELING AND SIMULATION IN DRUG DEVELOPMENT AND REGULATORY REVIEW 2913

Tab

le1.

New

Mol

ecu

lar

En

titi

esA

ppro

ved

inF

isca

lYea

r20

12(O

ctob

er20

11to

Sep

tem

ber

2012

)by

the

US

FD

A(f

rom

the

US

FD

AF

Y20

12In

nov

ativ

eD

rug

App

rova

lsR

epor

t:h

ttp:

//ww

w.f

da.g

ov/d

own

load

s/A

bou

tFD

A/R

epor

tsM

anu

alsF

orm

s/R

epor

ts/U

CM

3308

59.p

df)

Dru

gN

ame

App

rova

lDat

eIn

dica

tion

Orp

han

Dru

gA

ppro

ved

Fir

stin

US

AP

DU

FA

Dat

eM

etA

ppro

ved

Fir

stC

ycle

Spo

nso

r

I.P

rior

ity

Dru

gsA

MY

VID

(flor

beta

pir

F18

)A

pril

6,20

12T

oes

tim

ate

beta

-am

yloi

dpl

aqu

ede

nsi

tyin

brai

ns

ofpa

tien

tsw

ith

cogn

itiv

eim

pair

men

t

√√

Avi

dR

adio

-P

har

mac

euti

cals

,In

c.,P

hil

adel

phia

,P

enn

sylv

ania

CH

OL

INE

C11

Sep

tem

ber

12,2

012

For

PE

Tim

agin

gof

susp

ecte

dpr

osta

teca

nce

rh

ceN

A√

√M

ayo

Cli

nic

PE

TR

adio

chem

istr

yFa

cili

ty,

Roc

hes

ter,

Mic

hig

anE

RIV

ED

GE

(vis

mod

egib

)Ja

nu

ary

30,2

012

For

adva

nce

dba

salc

ellc

arci

nom

a√

√√

Gen

ente

ch,I

nc.

,Sou

thS

anF

ran

cisc

o,C

alif

orn

iaE

RW

INA

ZE

(asp

arag

inas

eer

win

iach

rysa

nth

emi)

Nov

embe

r18

,201

1F

orpa

tien

tsw

ith

acu

tely

mph

obla

stic

leu

kem

ia(A

LL

)an

dal

lerg

yto

E.c

oli-

deri

ved

aspa

ragi

nas

ean

dpe

gasp

arga

sech

emot

her

apy

dru

gs

√√

EU

SA

Ph

arm

a(U

SA

),In

c.,

Lan

ghor

ne,

Pen

nsy

lvan

ia

EY

LE

A(a

flib

erce

pt)

Nov

embe

r18

,201

1F

orw

et,a

ge-r

elat

edm

acu

lar

dege

ner

atio

n

√√

√R

egen

eron

Ph

arm

aceu

tica

ls,

Inc.

,Tar

ryto

wn

,New

York

JAK

AF

I(r

uxo

liti

nib

)N

ovem

ber

16,2

011

For

the

bon

em

arro

wdi

seas

em

yelo

fibr

osis

√√

√√

Incy

teC

orpo

rati

on,

Wil

min

gton

,Del

awar

eK

ALY

DE

CO

(iva

caft

or)

Nov

embe

r31

,201

1F

orcy

stic

fibr

osis

pati

ents

wit

hG

551D

mu

tati

on

√√

√√

Ver

tex

Ph

arm

aceu

tica

ls,I

nc.

,C

ambr

idge

,Mas

sach

use

tts

PE

RJE

TA

(per

tuzu

mab

)Ju

ne

8,20

12F

orH

ER

2-po

siti

vem

etas

tati

cbr

east

can

cer

√√

√G

enen

tech

,In

c.,S

outh

San

Fra

nci

sco,

Cal

ifor

nia

ST

IVA

RG

A(r

egor

afen

ib)

Sep

tem

ber

27,2

012

For

met

asta

tic

colo

rect

alca

nce

r√

√√

Bay

erH

ealt

hca

reP

har

mac

euti

cals

,In

c.,

Way

ne,

New

Jers

eyV

OR

AX

AZ

E(g

luca

rpid

ase)

Jan

uar

y17

,201

2T

otr

eat

toxi

cm

eth

otre

xate

con

cen

trat

ion

sin

plas

ma

ofpa

tien

tsre

ceiv

ing

chem

oth

erap

y

√√

√√

Btg

inte

rnat

ion

al,I

nc.

,Wes

tC

onsh

ohoc

ken

,P

enn

sylv

ania

XT

AN

DI

(en

zalu

tam

ide)

Au

gust

3,20

12F

orm

etas

tati

c,ca

stra

tion

-re

sist

ant

pros

tate

can

cer

√√

√M

ediv

atio

n,I

nc.

,Sou

thS

anF

ran

cisc

o,C

alif

orn

iaZ

ALT

RA

P(z

iv-a

flib

erce

pt)

Au

gust

3,20

12F

orm

etas

tati

cco

lore

ctal

can

cer

√√

√S

anofi

-Ave

nti

sU

.S.,

llc,

Bri

dgew

ater

,New

Jers

eyII

.Sta

nd

ard

Dru

gsA

UB

AG

IO(t

erifl

un

omid

e)S

epte

mbe

r12

,201

2F

orre

laps

ing

form

sof

mu

ltip

lesc

lero

sis

(MS

)

√√

√S

anofi

-Ave

nti

s,U

.S.,

llc.

BE

lVIQ

(lor

case

rin

hyd

roch

lori

de)

Jun

e27

,201

2F

orch

ron

icw

eigh

tm

anag

emen

t√

√A

ren

aP

har

mac

euti

cals

,In

c.,

Zofi

nge

n,S

wit

zerl

and

BO

SU

LIF

(bos

uti

nib

)S

epte

mbe

r4,

2012

For

chro

nic

mye

loge

nou

sle

uke

mia

(CM

L)

√√

√√

Pfi

zer

Inc.

,New

York

Cit

y,N

ewYo

rkE

lElY

SO

(tal

iglu

cera

seal

fa)

May

1,20

12F

orty

pe-1

gau

cher

dise

ase,

ara

rege

net

icdi

sord

er

√√

√P

rota

lix

Bio

ther

apeu

tics

Inc.

,C

arm

iel,

Isra

el

con

tin

ued

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Page 3: Huang 2013

2914 HUANG ET AL.

Tab

le1.

Con

tin

ued

.

Dru

gN

ame

App

rova

lDat

eIn

dica

tion

Orp

han

Dru

gA

ppro

ved

Fir

stin

US

AP

DU

FA

Dat

eM

etA

ppro

ved

Fir

stC

ycle

Spo

nso

r

FE

RR

IPR

OX

(def

erip

ron

e)O

ctob

er14

,201

1F

orir

onov

erlo

adin

pati

ents

wit

hth

alas

sem

ia(a

gen

etic

diso

rder

cau

sin

gan

emia

)

√√

Apo

phar

ma,

Inc.

,Tor

onto

,C

anad

a

GIN

TU

IT(a

llog

enei

ccu

ltu

red

kera

tin

ocyt

esan

dfi

brob

last

sin

bovi

ne

coll

agen

)

Mar

ch9,

2012

For

appl

icat

ion

tova

scu

lar

wou

nd

beds

inth

etr

eatm

ent

ofm

uco

gin

giva

lcon

diti

ons

√√

√O

rgan

ogen

esis

,In

c.,C

anto

n,

Mas

sach

use

tts

HE

MA

CO

RD

(hem

apoi

etic

prog

enit

orce

lls,

cord

(HP

C-C

)

Nov

embe

r10

,201

1F

oru

sein

un

rela

ted

don

orh

emat

opoi

etic

prog

enit

orce

lltr

ansp

lan

tati

onin

pati

ents

wit

hce

rtai

nbl

ood

diso

rder

s

NA

√√

New

York

Blo

odC

ente

r,In

c.,

New

York

Cit

y,N

ewYo

rk

HP

C,C

ord

Blo

od(h

emap

oiet

icpr

ogen

itor

cell

s,co

rd(H

PC

-C)

May

24,2

012

For

use

inu

nre

late

ddo

nor

hem

atop

oiet

icpr

ogen

itor

cell

tran

spla

nta

tion

inpa

tien

tsw

ith

cert

ain

bloo

ddi

sord

ers

NA

√√

Cli

nim

mu

ne

Lab

s,A

uro

ra,

Col

orad

o

INLY

TA

(axi

tin

ib)

Jan

uar

y27

,201

2F

orad

van

ced

kidn

eyca

nce

r√

√√

Pfi

zer,

Inc.

,New

York

Cit

y,N

ewYo

rkK

YP

RO

LIS

(car

filz

omib

)Ju

ly20

,201

2F

orm

ult

iple

mye

lom

a√

√√

√O

nyx

Ph

arm

aceu

tica

ls,I

nc.

,S

outh

San

Fra

nci

sco,

Cal

ifor

nia

LIN

ZE

SS

(lin

aclo

tide

)A

ugu

st30

,201

2F

orir

rita

ble

bow

elsy

ndr

ome

wit

hco

nst

ipat

ion

(IB

S-C

)

√√

√F

ores

tL

abor

ator

ies,

Inc.

,St.

Lou

is,M

isso

uri

ME

NH

IBR

IX(M

enin

goco

ccal

Gro

ups

Can

dY

and

Hae

mop

hil

us

bT

etan

us

Tox

oid

Con

juga

teV

acci

ne)

Jun

e14

,201

2C

ombi

nat

ion

vacc

ine

topr

even

tm

enin

goco

ccal

dise

ase

and

Hae

mop

hil

us

infl

uen

zae

type

b(H

ib)

inch

ildr

en

√√

Gla

xoS

mit

hK

lin

eB

iolo

gica

ls,

base

din

Rix

ensa

rt,

Bel

giu

m

MY

RB

ER

TIQ

(mir

abeg

ron

)Ju

ne

28,2

012

For

trea

tmen

tof

over

acti

vebl

adde

r√

√A

stel

las

Ph

arm

aG

loba

lD

evel

opm

ent,

Inc.

,N

orth

broo

k,Il

lin

ois

NE

UT

RO

VA

L(t

bo-fi

lgra

stim

)A

ugu

st29

,201

2T

ore

duce

dura

tion

ofn

eutr

open

iain

chem

oth

erap

ypa

tien

ts

√S

icor

Bio

tech

UA

BV

iln

ius,

Lit

hu

ania

OM

ON

TY

S(p

egin

esat

ide)

Mar

ch27

,201

2F

oran

emia

inch

ron

icki

dney

dise

ase

pati

ents

ondi

alys

is

√√

√A

ffym

ax,I

nc.

,Pal

oA

lto,

Cal

ifor

nia

ON

FI

(clo

baza

m)

Oct

ober

21,2

011

For

seiz

ure

sas

soci

ated

wit

hL

enn

ox-G

asta

ux

syn

drom

e

√√

√L

un

dbec

k,In

c.,D

eerfi

eld,

Illi

noi

sP

ICA

TO

(in

gen

olm

ebu

tate

)Ja

nu

ary

23,2

012

For

the

topi

calt

reat

men

tof

acti

nic

kera

tosi

s

√√

√L

eoP

har

ma

AS,

Bal

leru

p,D

enm

ark

PR

EP

OP

IK(s

odiu

mpi

cosu

lfat

e,m

agn

esiu

mox

ide,

citr

icac

id)

July

16,2

012

For

colo

ncl

ean

sin

gin

prep

arat

ion

for

colo

nos

copy

√√

Fer

rin

gP

har

mac

euti

cals

Par

sipp

any,

New

Jers

ey

con

tin

ued

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013 DOI 10.1002/jps

Page 4: Huang 2013

THE UTILITY OF MODELING AND SIMULATION IN DRUG DEVELOPMENT AND REGULATORY REVIEW 2915

Tab

le1.

Con

tin

ued

.

Dru

gN

ame

App

rova

lDat

eIn

dica

tion

Orp

han

Dru

gA

ppro

ved

Fir

stin

US

AP

DU

FA

Dat

eM

etA

ppro

ved

Fir

stC

ycle

Spo

nso

r

ST

EN

DR

A(a

van

afil)

Apr

il27

,201

2F

orth

etr

eatm

ent

ofer

ecti

ledy

sfu

nct

ion

(ED

)

√√

Viv

us

Inc.

,Mou

nta

inV

iew

,C

alif

orn

iaS

TR

IBIL

D(e

lvit

egra

vir,

cobi

cist

at,e

mtr

icit

abin

e,te

nof

ovir

diso

prox

ilfu

mar

ate)

Au

gust

27,2

012

For

trea

tmen

tof

HIV

-1√

√√

Gil

ead

Sci

ence

s,In

c.,F

oste

rC

ity,

Cal

ifor

nia

SU

RF

AX

IN(l

uci

nac

tan

t)M

arch

6,20

12T

opr

even

tre

spir

ator

ydi

stre

sssy

ndr

ome

(RD

S)

inpr

emat

ure

infa

nts

√√

Dis

cove

ryL

abor

ator

ies,

Inc.

,W

arri

ngt

on,P

enn

sylv

ania

TU

DO

RZ

AP

RE

SS

AIR

(acl

idin

ium

brom

ide)

July

23,2

012

For

lon

g-te

rmm

ain

ten

ance

trea

tmen

tof

bron

chos

pasm

inC

OP

D

√√

√F

ores

tL

abor

ator

ies,

Inc.

,St.

Lou

is,M

isso

uri

ZIO

PT

AN

(tafl

upr

ost)

Feb

ruar

y10

,201

2T

ore

duce

intr

aocu

lar

pres

sure

(IO

P)

inpa

tien

tsw

ith

glau

com

aor

ocu

lar

hyp

erte

nsi

on

√M

erck

Sh

arp

and

Doh

me

Cor

pora

tion

,Wh

iteh

ouse

Sta

tion

,New

Jers

ey

(2) the potential for physiologically based pharma-cokinetic (PBPK) modeling to address questions re-lated to individualization of doses based on patient-specific factors (e.g., age, sex, race, organ dysfunc-tion, concomitant drug administration), and (3) re-quired processes for the development of predictivesafety tools employing preclinical, in vitro, and in sil-ico methods using the vast US FDA database andother “precompetitive” information with collabora-tions from multiple internal and external parties.

PHARMACOMETRIC MODELING

The utility of modeling and simulation in drugdevelopment and regulatory review has been welldocumented.10,12 Among the many applications, mostof the OCP efforts have been focused on late phasedecisions such as dosing and labeling or postapprovalevaluation of unstudied doses or dosing regimens.10,11

Recently, we have increased efforts to apply modelingand simulation to assist in dose selection and trialdesign for late phase trials based on data obtainedfrom early phase clinical trials. Three case studiesare highlighted below to illustrate the application ofpharmacometric analyses to address two major chal-lenges: dose optimization and extrapolation of thera-peutic effect to unstudied populations.13–18 The mod-els applied in our evaluation are listed in Table 2.

Dose Optimization—Experience from Trastuzumab forMetastatic Gastric Cancer

Dose optimization has not been the focus of oncol-ogy drug development until recent years. Insufficientselection of dose and dosing schedule is one of themany reasons that could be related to the low suc-cess rates in oncology drug development programsin which dose-ranging trials to optimize dose selec-tion for efficacy are rarely conducted. In the absenceof activity-based, dose-ranging studies, exposure–re-sponse (E–R) analyses of data from “pivotal” efficacytrials may aid in dose refinement and/or risk/benefitevaluation; here, we present one such example.

Trastuzumab was initially approved by the USFDA in 1998 for metastatic HER2-overexpressingbreast cancer. In 2010, the US FDA approvedtrastuzumab for HER2-overexpressing metastaticgastric cancer (mGC). In the phase 3 trial fortrastuzumab for mGC, the same dosing regimen ap-proved for breast cancer was chosen for the new pa-tient population. During the regulatory review, ananalysis based on clinical data obtained from thisphase 3 trial, which employed one dose level, wasconducted to assess whether the proposed dosing reg-imen was supported by the E–R relationship. Be-cause of the nonrandomized nature of this E–R anal-ysis, there were multiple confounding factors. To ad-dress this, an additional case–control analysis was

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Page 5: Huang 2013

2916 HUANG ET AL.

Table 2. Models Used in Three Cases Presented Under the “Pharmacometric Modeling” Section

Trastuzumab Boceprevir/telaprevir Topiramate

Model Compartmental pharmacokinetic model for exposureand statistical models (Cox model and case–controlanalysis) for exposure–response analysis

Mathematical models for proportionsin subgroups of a population

Compartmentalpharmacokinetic model

conducted to minimize the bias in the estimationof the treatment effect at a certain exposure level.The matched factors included Eastern CooperativeOncology Group Performance Score, prior gastrec-tomy, number of metastatic sites, Asian ethnicity, andimmunohistochemistry.13 The pharmacometric anal-ysis suggested that patients in the lowest quartileof trastuzumab trough concentrations were at signif-icant risk of treatment failure (as measured by de-creased overall survival). These patients could be sub-ject to known serious toxicities of trastuzumb withoutthe benefits from the trastuzumab treatment. As a re-sult of the pharmacometric analysis, several postmar-keting requirement (PMR) studies were requested ofthe sponsor to conduct further evaluations, includingone trial to evaluate alternate (higher or individual-ized) dosing regimens.14 This type of pharmacometricanalysis, if conducted early during drug development,may help optimize patient dose and improve the suc-cess rate of phase 3 trials of oncology drug products.

Extrapolation of Indications to Unstudied PatientGroup—Boceprevir and Telaprevir for ChronicHepatitis C

Chronic hepatitis C is a major cause of cirrhosis inNorth America. Before the US FDA’s approval of bo-ceprevir and telaprevir in May 2011, the standard ofcare for chronic hepatitis C was peginterferon alfain combination with ribavirin for 48 weeks. Tradi-tionally, patients with chronic hepatitis C are dividedinto two broad categories (treatment naive and treat-ment experienced) based on their previous exposureto interferon-based therapy. Treatment experiencedsubjects who failed the previous round of peginter-feron/ribavirin treatment are further classified intorelapsers, partial responders, and null responders.

Boceprevir and telaprevir are hepatitis C virus(HCV) NS3/4A protease inhibitors and represent anew class of small molecules that directly targetsvirus replication. Despite the convincing efficacy re-sults of both drugs in treatment-naive patients, therewere still considerable concerns about the efficacyand the optimal doses in the treatment-experiencedand a subgroup of treatment-naive patients.15 Un-resolved questions addressed in the boceprevir andtelaprevir reviews included: (1) Is there evidence ofboceprevir effectiveness for prior null responders withpeginterferon/ribavirin (a subgroup that was specifi-cally excluded from the pivotal study)? (2) What is

the appropriate boceprevir dosing regimen for a sub-set of treatment-naive subjects who are late respon-ders?, and (3) Can telaprevir response guided ther-apy, which can shorten the treatment duration from48 weeks to 24 weeks in >60% of the treatment-naivepatients, be approved in prior relapsers with pegin-terferon/ribavirin (a group in which response guidedtherapy was not prospectively evaluated). To addressthese challenging questions, multiple additional clin-ical trials would typically be required by US FDA togather additional information. However, a novel phar-macometric method16,17 was applied in the reviewprocess to quantitatively bridge knowledge betweentreatment naive and experienced patients when in-formation was lacking in one population. Detaileddescription of the method is described in separatepapers.16,17 This analysis demonstrated that inter-feron responsiveness for a second course of pegin-terferon/ribavirin treatment did not change after thefirst course of treatment. This novel pharmacometricmethod was presented at both boceprevir and telapre-vir Advisory Committee meetings and the analysesserved as the foundation for the following US FDAdecisions on telaprevir and boceprevir: (1) the effec-tiveness of boceprevir for the unstudied treatment-experienced subgroup can be established from avail-able data in treatment-naive patients; (2) the ap-propriate boceprevir dosing regimen for treatment-naive subjects who are late responders was estab-lished based on data from treatment-experienced pa-tients; and (3) telaprevir response guided therapy canbe approved in a subgroup that was not studied in thetrials. The approval of these two new drugs for HCVpatients with appropriate dosing regimens providedthe timely benefit to all HCV patients and avoideddelay and suboptimal dosing regimen in certain sub-groups.

Extrapolation of Indications to Specific PediatricPatients—Topiramate for Epilepsy

Anticonvulsants are generally studied and approvedinitially for adjunctive use as such clinical efficacytrials can be readily performed. However, subsequentclinical trials for monotherapy present particular ex-perimental and ethical challenges, especially for pe-diatric patients. Therefore, alternative methods areneeded to substantiate the approval decision and toidentify the appropriate dosing regimen for monother-apy in pediatrics.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013 DOI 10.1002/jps

Page 6: Huang 2013

THE UTILITY OF MODELING AND SIMULATION IN DRUG DEVELOPMENT AND REGULATORY REVIEW 2917

Topiramate has been approved for adjunctive ther-apy in adults and in children 2 years and older basedon phase 3 trials. It is also approved for monotherapyto treat partial onset seizures and primary general-ized tonic–clonic seizures in adults and children 10years and older based on phase 3 trials. Efficacy datafor monotherapy in pediatrics 2–10 years old could notbe obtained from clinical efficacy trials. A pharmaco-metric method was developed and applied to quanti-tatively bridge knowledge between adults and pedi-atrics in monotherapy and adjunctive therapies. Thisapproach consisted of first showing a similar E–R re-lationship between adults and pediatric patients 2years and older when topiramate was given as an ad-junctive therapy. Next, the similarity of the E–R re-lationships was demonstrated in adults and pediatricpatients ages 6 to <16 years when topiramate wasgiven as initial monotherapy. Specific dosing in chil-dren 2 to <10 years of age was then derived from sim-ulations utilizing plasma exposure ranges observed inpediatric and adult patients treated with topiramatein monotherapy. The above analyses made it possi-ble to extrapolate monotherapy efficacy from patients10 years and older to pediatric patients 2 to <10 yearsold by matching the topiramate exposures in thesepediatrics to exposures observed in adults and olderpediatric patients. The safety data obtained in the rel-evant pediatric patients given at a higher dose level oftopiramate was used to ensure that the targeted topi-ramate exposure is within the “safe” exposure range.Because of the limited strengths of topiramate tablet(the lowest strength is 25 mg), an additional optimiza-tion algorithm was implemented to individualize thedosing regimen to target the desired topiramate expo-sure as closely as possible for each pediatric patient.This individualized dosing regimen was approved fortopiramate pediatric monotherapy.18

PBPK MODELING

Physiologically based pharmacokinetic modeling hasbeen used extensively in the past in the estimation ofhuman exposure of environmental chemicals. Recentadvancement in the understanding of physiologicaland biological processes, drug disposition includingdrug transport,19 and computer science, coupled withthe availability of several specialized PBPK softwarepackages, has contributed to more widespread useof PBPK in drug discovery, drug development, andregulatory review. PBPK has been applied for earlyin vitro to in vivo predictions of the PK of investiga-tional drugs in first-in-human studies20,21 and for theevaluation of the effects of intrinsic and extrinsic fac-tors, alone or in combination, on drug exposure.22,23

Between 2008 and 2012, the US FDA received 33IND/NDA (Investigational New Drug/New Drug Ap-plication) submissions containing PBPK modeling ap-proaches. In parallel with the increasing number ofsubmissions containing PBPK, the agency has in-creasingly utilized de novo (i.e., US FDA initiated)PBPK in its reviews to help characterize PK in avariety of complex clinical scenarios. Figure 1 showsthe number of submitted IND/NDA containing PBPK,and the number of models developed by OCP duringits regulatory review from 2008 to 2012. The followingcases illustrate application of PBPK to inform severaldecisions related to therapeutic use from a regulatorystandpoint (Fig. 2).

Drug–Drug Interactions

Case 1—Focused Clinical Drug Interaction StudiesBased on in Vitro Inhibition Data

Cabazitaxel is approved for hormone-refractorymetastatic prostate cancer. Although in vitro datasuggested that it could inhibit CYP3A, a PBPK

Figure 1. The number of PBPK applications contained in IND/NDA submissions or developedby US FDA reviewers from 2008 to 2012.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Page 7: Huang 2013

2918 HUANG ET AL.

Figure 2. Areas of applications in the 33 PBPK submissions in IND/NDA received by USFDA’s Office of Clinical Pharmacology from 2008 to 2012.

analysis, considering the drug’s disposition (intra-venous administration, short plasma half-life, etc.),indicated minimal effect on the exposure of midazo-lam (a CYP3A probe) in humans, even at an inhi-bition potency that is an order of magnitude higherthan that obtained in vitro. The simulation resultsobviated the need for the US FDA review team torequest a PMR study to evaluate a possible interac-tion between cabazitaxel and CYP3A substrates inhumans.23

Case 2—Extrapolation to Unstudied Drug InteractionScenarios

Fesoterodine is a prodrug of 5-hydroxymethyl toltero-dine (5-HMT) for the treatment of overactive blad-der. The recommended initial dose is 4 mg daily,which can be increased to 8 mg once daily. The ac-tive moiety 5-HMT is significantly metabolized byCYP3A4 and polymorphic CYP2D6, in addition toundergoing renal excretion. Several drug interactionand pharmacogenetic studies have been conducted inCYP2D6 extensive metabolizers (EM) and poor me-tabolizers (PM).24,25 The results are summarized inTable 3. On the basis of these data, the labeling indi-cated that the recommended fesoterodine daily doseshould not exceed 4 mg when taken with a potentCYP3A inhibitor, such as ketoconazole, itraconazole,and clarithromycin.26 However, whether fesoterodinedose needs to be adjusted when coadministered witha moderate CYP3A inhibitor (e.g., fluconazole) in sub-jects with varying CYP2D6 activities, is the nextregulatory question. A PBPK model of 5-HMT wasconstructed using in vitro metabolism and humanPK data. The model was verified with the availabledrug–drug interaction and pharmacogenetic data. Asshown in Table 3, the model-simulated fold changes inAUC and Cmax under various scenarios that have beentested in humans were in close agreement with thoseobserved. Compared with CYP2D6 EM without tak-ing fluconazole, the PBPK simulated fold-changes inthe AUC of 5-HMT were 1.3 and 2.6-fold, respectively,when fluconazole was coadministered in CYP2D6 EM

Table 3. Observed and PBPK-Simulated 5-HMT ExposureChanges, Measured by the AUC Ratio of 5-HMT, in the Presenceof Strong and Moderate CYP3A4 Inhibitors in Subjects withCYP2D6 Extensive (EM) or Poor Metabolizer (PM) Status

AUC Ratio of 5-HMT

Observed Predicted

EM with/without ketoconazole 2.3 1.9PM with/without ketoconazole 2.5 3.3PM/EM 2.3 1.6PM with ketoconazole/EM without

ketoconazole5.7 5.4

EM with/without fluconazole 1.3 1.3PM with fluconazole/EM without

fluconazole– 2.6

and PM, respectively. The model simulated exposuredata of 5-HMT in CYP2D6 PMs taking a moderateCYP3A inhibitor, along with known data in CYP2D6EMs taking fluconazole, provided the needed infor-mation for regulatory review and decision making.27

The current labeling indicated that “there is no clini-cally relevant effect of moderate CYP3A inhibitors onthe pharmacokinetics of fesoterodine.”26

Application to Specific Patient Populations–-Pediatrics

Physiologically based pharmacokinetic models havebeen developed and applied in pediatric drugdevelopment.28–30 These pediatric PBPK modelsleverage extensive drug-independent information de-scribing growth- and development-related physiologi-cal processes in pediatric patients based on publisheddata.28–30 Between 2008 and 2012, OCP reviewed sixpediatric submissions containing PBPK, four of whichwere discussed in a recent report describing the ap-plication of PBPK in pediatric drug development.31

The models were used to simulate pharmacokinet-ics of investigational drugs in pediatric populationsof different age groups to support dose selection forclinical trials. For example, a PBPK model of a drugwas used to determine the dose for the 12–18-year-oldgroup that would produce systemic exposure levelsmatching those in adults. On the basis of the clinical

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013 DOI 10.1002/jps

Page 8: Huang 2013

THE UTILITY OF MODELING AND SIMULATION IN DRUG DEVELOPMENT AND REGULATORY REVIEW 2919

data obtained in this age group, the model was sub-sequently refined to determine the appropriate dosesfor younger age groups. Some of these pediatric PBPKsimulations also explored situations when a dedicatedclinical PK study is difficult to conduct. For exam-ple, for a recently approved progestin-containing in-trauterine system for prevention of pregnancy,32 thesponsor used PBPK to simulate plasma exposure oflevonorgestrel in postmenarcheal pediatric subjects(up to 18-year old). A physiological uterine compart-ment was incorporated into the PBPK model, whichallowed simulation of uterine drug release using ki-netic data determined in vitro. The simulated pedi-atric PK of levonorgestrel in pediatric subjects sup-ported the use of this product in a pediatric trial forfemales postmenarcheal to 18, an age group for whichthere is currently no data.

A recent US FDA Clinical Pharmacology AdvisoryCommittee meeting33 discussed the clinical pharma-cology aspects of pediatric clinical trial design anddosing to optimize pediatric drug development. Thecommittee endorsed the use of modeling and sim-ulation in pediatric drug development. Importantly,the committee emphasized that PBPK models needto be prospectively verified using adult PK data, in-cluding those from drug interactions, renal impair-ment, hepatic impairment available at the time ofmodel building.33 In addition, the committee empha-

sized the drug’s disposition needs to be well definedto yield maximally informative models-based results.

MECHANISM-BASED DRUG SAFETYEVALUATION USING SYSTEMSPHARMACOLOGY

As pharmacology and clinical pharmacology move for-ward from reductionist approaches toward integra-tive systems approaches to address problems, OCPhas initiated an effort to leverage the new sciencethat is evolving in systems pharmacology.34 This isfocused on the prediction of adverse drug events us-ing the tools of cheminformatics, bioinformatics, andsystems biology.35 To move regulatory science forwardin this area, the approach and the tools that still re-quire development have recently been defined.36

Ontology of Adverse Events

The initial activities are both within the US FDA andin collaboration with partners in the pharmaceuticalindustry and academia. The framework for a predic-tive drug safety systems model is being structured asa series of ontologies at different levels of biologicalorganization.37 These are developed to organize themassive amount of pertinent information that willpopulate this framework. (see Fig. 3) This includesan “ontology of adverse events” that is building on

Figure 3. Interaction map showing n number of drugs, k number of proteins, m number ofpathways, and h number of adverse drug reactions (ADRs). Abbreviation: GI, gastrointestinal(reproduced with permission from reference D3).

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Page 9: Huang 2013

2920 HUANG ET AL.

the ontology of adverse events recently developed forcharacterization of vaccine related adverse events.38

All of these ontologies are developed in the format con-sistent with recommendations from the National Cen-ters for Biomedical Ontology to make use of standard-ized elements in existing ontologies at various levelsof biological organization (e.g., gene ontology, cell lineontology, systematized nomenclature of medicine clin-ical terms).39 A major challenge that is currently be-ing addressed in this program within the US FDAis the aggregation and definition of MedDRA termsin a manner that allows these terms to be mappedto organ dysfunction, cell dysfunction, and the bio-logical pathways and alterations in gene expressionthat are associated with drug-induced toxicities. Asthis evolves, it will constitute the systems pharma-cology network that defines a drug mediated adverseevent from observed changes in gene expression upthrough organelle function, cellular expression, or-gan level toxicity and that ultimately maps to theMedDRA terms.

Databases—Leveraging “Precompetitive” Data

An important element of the program is the collec-tion and organization of drug toxicity data. Publisheddata are relatively easy to obtain and organize, and anumber of public and proprietary efforts are creatingsuch databases. These are useful for different formsof data mining and establishing associations betweendrug toxicities, the data supporting the mechanism ofsuch toxicity, and in some instances the prediction oftoxicity from similar chemical structures.40 In otherinstances the prediction of clinical toxicities based onthe biological pathways that are involved based ondata in the public domain for drugs that have simi-lar targets has been used.41 More challenging to ob-tain are the data contained in drug development pro-grams, either those that were successful, or equallyimportant, those that were not successful due to drugtoxicity or for other reasons. An effort is underway toencourage pharmaceutical companies to define whatof these data can be viewed as “precompetitive” andcan be shared to build more complete drug toxicitydatabases that are publicly available. All of thesedata will be placed in a common platform that al-lows easy integration across different data formats. Alikely platform for this will be tranSMART, a utilitydeveloped within the pharmaceutical industry, but re-cently placed in the public domain under a nonprofit501c3 format.42,43 We believe this will evolve into apublic drug safety data warehouse that will be mod-eled after other such data warehouses sponsored byUS FDA such as the EKG data warehouse.44 Goingforward such a resource will contain the data neces-sary to populate the systems networks that are de-fined for specific drug toxicities and their mechanism.In addition it is likely to be useful to the greater drug

development community to allow greater efficiencyfor drug candidate toxicity prediction and thus drugcandidate selection or patient population selection fora drug candidate. At present such databases reside inmany of the larger pharmaceutical companies, how-ever they are limited to the data that resides in thatcompany and to some extent augmented by data inthe public domain.

Initial Test Cases: Cardiotoxicity and Hepatotoxicity

The extent of the systems network needed to effec-tively predict particular drug toxicities is likely to bevariable depending on the target(s), both desired andnot desired, for the drug or series of related drugsto be evaluated. An initial “use” case is being devel-oped to study the non-QT cardiotoxicity of tyrosine ki-nase inhibitor drugs. The clinical reports of depressedleft ventricular cardiac function with exposure to se-lected tyrosine kinase inhibitors have certainly raiseda potential safety signal.45 However, the extent or re-versibility of this cardiotoxicity is not clear and it isalso unclear if this is a class effect or only tyrosinekinase inhibitors that target specific kinases are im-plicated. The predictive systems model for this case isbeing developed as a collaborative effort, primarily atthe Systems Biology Institute 46,47 and the Universityof Tokyo with the US FDA as a collaborative partner.A number of pharmaceutical companies are express-ing interest in joining the collaboration, as there isextensive effort to target various tyrosine kinases foran increasing number of clinical indications.

A systems network for the prediction of druginduced hepatotoxicity is being developed at TheHamner Institute in collaboration with a numberof pharmaceutical companies and with US FDA in-teracting and contributing when appropriate. Thisis a very highly specified model that encompassesa limited, but very well characterized, systems net-work. The utility of this approach from in vitro/in vivo extrapolation and across species hepatotoxiceffects for methapyrilene and acetaminophen wasdemonstrated.48 This is somewhat in contrast to othersystems analysis efforts such as the one at the Sys-tems Biology Institute, which cover a wider systemsnetwork, however with relationships between nodesin the system less well characterized. As experienceaccumulates, the comparative utility of these ap-proaches or a synthesis of them will become more evi-dent. At this time the extent of the overall system andhow well characterized the relations between nodesand edges of the system are necessary for a particu-lar predictive drug toxicity problem requires furtherexploration and testing.

Applications of this program to regulatory deci-sion making has to date been focused on the use ofdata mining to establish relationships between com-mon biological pathways across drugs and targets and

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013 DOI 10.1002/jps

Page 10: Huang 2013

THE UTILITY OF MODELING AND SIMULATION IN DRUG DEVELOPMENT AND REGULATORY REVIEW 2921

expected or observed clinical safety signals. In addi-tion the tools of cheminformatics have been incorpo-rated to predict toxicities based on molecular struc-tural elements of the drug or compound class in as-sociation with the biological pathways affected. Thislatter activity is a further evolution of the quanti-tative structure–activity relationship effort that hasbeen ongoing at US FDA for some years.49 The out-comes have been incorporation of added safety infor-mation in labeling in some cases, providing a basisto not include certain risks in other cases, and signalstrengthening and in some cases signal weakeningbased on pharmacological mechanistic rationale forpotential safety signals noted in post market adverseevent monitoring.

CONCLUSION AND FUTURE DIRECTIONS

United States Food and Drug Administration hasbeen proactive in the development of regulatory sci-ence to address an array of public health challenges.Modeling and simulation have been important scien-tific investment areas for US FDA’s OCP, and willcontinue to be a major area of growth. Modeling andsimulation, tools of quantitative and systems phar-macology, will need to be put into the larger trans-lational science context to reach full potential. Keyissues will need to be addressed (probably in a recur-ring fashion) to see enhanced uptake of modeling andsimulation approaches in drug regulation.50–52 Keyneeds include:

• Better understanding of the mechanisms ofdrug action, including off target effects and max-imal elucidation of the disposition pathways ofdrug molecules

• “Vertical integration”: synergistic assimilationof the bottom up (cellular level) and the top-down (organism level) models to scale frommolecular interactions to organismal physiology

• Targeted training and education of regulatoryand nonregulatory scientists

• Sharing of precompetitive data (preclinicaland clinical datasets); efficient use of theprivate–public partnership models to fosteracademia–industry–government interactions

• Development of best practices in the develop-ment of models fit for regulatory use

• Development of mechanisms that allow fortimely evaluation (and reevaluation) of modelsin view of rapidly evolving methodologies andscience

• Development of robust PD markers to facilitatethe continuing development in the E–R relation-ship

Despite these challenges, improved understandingof molecular mechanisms is enabling us to employmodeling and simulation in evaluation of “subset” ef-

fects (based on subtype of diseases, age, sex, race, ge-netics, organ dysfunctions, concomitant medications,etc.). For example, recently published US FDA guid-ance documents related to drug interactions53 andearly phase pharmacogenomic evaluation54 have in-cluded recommendations for the use of PBPK whereappropriate. There are several US FDA and ICHguidelines that discuss the relevance of modeling inseveral aspects of drug development.51,55,56 Meaning-ful, pragmatic advice to drug developers will needto be science and experience based. As such, theknowledge gained from predict-learn-confirm exer-cises will contribute to regulatory decision mak-ing, and collaboration among stakeholders—industry,global regulatory agencies, academia, and others willbe important.2,4,6,57,58

REFERENCES

1. US FDA innovative drug approvals. Accessed, at: http://www.fda.gov/AboutFDA/ReportsManualsForms/Reports/ucm276385.htm.

2. Honig PK, Huang S-M. 2012 March. Regulatory science andthe role of the regulator in biomedical innovation. Clin Phar-macol Ther 91(3):347–352.

3. US FDA Safety and Innovation Act (FDASIA). Ac-cessed, at: http://www.fda.gov/RegulatoryInformation/Legislation/FederalFoodDrugandCosmeticActFDCAct/SignificantAmendmentstotheFDCAct/FDASIA/ucm20027187.htm.

4. Buckman S, Huang S-M, Murphy S. 2007. Medical productdevelopment and regulatory science for the 21st century: Thecritical path vision and its impact on health care. Clin Phar-macol Ther 81:141–144.

5. Woodcock J, Woosley R. 2008. The FDA critical path initiativeand its influence on new drug development. Annu Rev Med59:1–12.

6. Barratt RA, Bowens SL, McCune SK, Johannessen JN, Buck-man SY. February 2012. The critical path initiative: Lever-aging collaborations to enhance regulatory science. Clin Phar-macol Ther 91(3):380–383

7. Advancing Regulatory Science for Public Health—A Frame-work for FDA’s Regulatory Science Initiative, October2010. Accessed February 26, 2013, at: http://www.fda.gov/downloads/ScienceResearch/SpecialTopics/RegulatoryScience/UCM228444.pdf.

8. Bashaw ED, Huang S-M, Cote TR, Pariser AR, Garnett CE,Burckart G, Zhang L, Men AY, Le CD, Charlab R, GobburuJV, Lesko LJ. November 2011. Clinical pharmacology as acorner stone of orphan drug development. Nat Rev Drug Discov10:795–796.

9. Florian J, Garnett CE, Nallani SC, Rappaport BA, Throck-morton DC. 2012. A modeling and simulation approach tocharacterize methadone QT prolongation using pooled datafrom five clinical trials in MMT patients. Clin Pharmacol Ther91:666–672.

10. Lee JY, Garnett CE, Gobburu JV, Bhattaram VA, Brar S, EarpJC, Jadhav PR, Krudys K, Lesko LJ, Li F, Liu J, MAdabushiR, Marathe A, Nehrotra N, Tornoe C, Wang Y, Zhu H. 2011.Impact of pharmacometric analyses on new drug approval andlabeling decisions: A review of 198 submissions between 2000and 2008. Clin Pharmacokinet 50(10):627–635.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013

Page 11: Huang 2013

2922 HUANG ET AL.

11. Huang S-M, Bhattaram A, Mehrotra N, Wang Y. 2013. Is thisthe dose for you?—The role of modeling. Clin Pharmacol Ther93:159–162.

12. Lalonde RL, Kowalski KG, Hutmaher MM, Ewy W, NicholsDJ, Milligan PA, Corrigan BW, Lockwood PA, Marshall SA,Benincosa LJ, Tensfeldt TG, Parivar K, Amantea M, Glue P,Loide H, Miller R. 2007. Model-based drug development, ClinPharmacol Ther 82:21–32.

13. Yang J, Zhao H, Garnett C, Rahman A, Gobburu JV, Pierce W,Schechter G, Summers J, Keegan P, Booth B, Wang Y. 2013.Combination of exposure–response and case–control analysesin regulatory decision making. J Clin Pharmacol 53:160-6.

14. Postmarket requirement studies for trastuzumab. Accessed,at: http://www.accessdata.fda.gov/drugsatfda docs/appletter/2010/103792s5250ltr.pdf.

15. US FDA Antiviral Advisory Committee meeting on bo-ceprevir and telapravir, April 27 and April 28, 2011,Accessed January 28, 2013, at: http://www.fda.gov/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/AntiviralDrugsAdvisoryCommittee/ucm247236.htm.

16. Liu J, Jadhav PR, Amur S, Fleischer R, Hammerstrom T,Lewis L, Naeger L, O’Rear J, Pacanowski M, Robertson S,Seo S, Soon G, Birnkrant D. 2012. Response guided telapre-vir therapy in prior relapsers?: The role of bridging datafrom treatment-naive and experienced subjects. Hepatology57:897–902.

17. Florian J Jr., Jadhav PR, Amur S, Ayala R, Harrington P,Mishra P, Pacanowski M, Robertson S, Singer M, Soon G, ZengW, Murray J. 2012. Boceprevir dosing for late responders andnull responders: The role of bridging data between treatment-naive and experienced subjects, Hepatology 57:903–907.

18. Topiramate labeling. Accessed December 30, 2012, at:http://www.accessdata.fda.gov/drugsatfda docs/label/2011/020505s042,020844s036lbl.pdf.

19. The International Transporter Consortium, ITC (GiacominiKM, Huang S-M, Tweedie DJ, Benet LZ, Brouwer KL, Chu X,Dahlin A, Evers R, Fischer V, Hillgren KM, Hoffmaster KA,Ishikawa T, Keppler D, Kim RB, Lee CA, Niemi M, Polli JW,Sugiyama Y, Swaan PW, Ware JA, Wright SH, Yee SW, Zamek-Gliszczynski MJ, Zhang L). 2010. Membrane transporters indrug development: Report form the FDA critical path initiativesponsored workshop. Nat Rev Drug Discov 9:215–236.

20. Rowland M, Peck C, Tucker G. 2011. Physiologically-basedpharmacokinetics in drug development and regulatory science.Annu Rev Pharmacol Toxicol 51:45–73.

21. Rostami-Hodjegan A. 2012. Physiologically based pharma-cokinetics joined with in vitro–in vivo extrapolation of ADME:A marriage under the arch of systems pharmacology. ClinPharmacol Ther 92:50–61.

22. Zhao P, Zhang L, Grillo JA, Liu Q, Bullock JM, Moon YJ, SongP, Brar SS, Madabushi R, Wu TC, Booth BP, Rahman NA,Reynolds KS, Gil BE, Lesko LJ, Huang SM. 2011. Applica-tions of physiologically based pharmacokinetic (PBPK) model-ing and simulation during regulatory review. Clin PharmacolTher 89:259–267.

23. Huang SM, Rowland M. 2012. The role of physiologically basedpharmacokinetic modeling in regulatory review. Clin Pharma-col Ther 91:542–549.

24. Malhotra B, Dickins M, Alvey C, Jumadilova Z, Li X, Duczyn-ski G, Gandelman K. 2011. Effects of the moderate CYP3A4inhibitor, fluconazole, on the pharmacokinetics of fesoterodinein healthy subjects. Br J Clin Pharmacol 72(2):263–269.

25. Fesoterodine clinical pharmacology and biopharmaceutics re-view. Accessed December 31, 2012, at: http://www.accessdata.fda.gov/drugsatfda docs/nda/2008/022030s000 ClinPharmR.pdf.

26. US FDA labeling-Toviaz (fesoterodine fumarate) for oraladministration. August 2012. Accessed January 29, 2013,

at: http://www.accessdata.fda.gov/drugsatfda docs/label/2012/022030s009lbl.pdf.

27. Vieira ML, Zhao P, Kim M, Apparaju S, Huang S. 2012. Pre-dicting CYP3A4 inhibition in CYP2D6 poor metabolizers usingPBPK modeling and simulation: Fesoterodine as an example.Clin Pharmcol Ther 91S1 S53.

28. Edginton AN, Schmitt W, Willmann S. 2006. Development andevaluation of a generic physiologically based pharmacokineticmodel for children, Clin Pharmacokinet 45:1013–1034.

29. Johnson TN, Rostami-Hodjegan A. 2011. Resurgence in theuse of physiologically based pharmacokinetic models in pedi-atric clinical pharmacology: Parallel shift in incorporating theknowledge of biological elements and increased applicabilityto drug development and clinical practice. Paediatr Anaesth21:291–301.

30. Barrett JS, Alberighi ODC, Laer S, Meibohm B. June 2012.Physiologically based pharmacokinetic (PBPK) modeling inchildren. Clin Pharmacol Ther 92:40–49.

31. Leong R, Vieira ML, Zhao P, Mulugeta Y, Lee CS, HuangSM, Burckart GJ. 2012. Regulatory experience with physio-logically based pharmacokinetic modeling for pediatric drugtrials. Clin Pharmacol Ther 91:926–931.

32. US FDA label and review on Skyla (levonorgestrel-releasingintrauterine system), drugs at the FDA. Accessed January30, 2013, at: http://www.accessdata.fda.gov/Scripts/cder/DrugsatFDA/index.cfm?fuseaction=Search.Label_ApprovalHistory#apphist.

33. US FDA Pharmaceutical Science and Clinical PharmacologyAdvisory Committee meeting, March 14, 2012. Accessed Jan-uary 30, 2013, at: http://www.fda.gov/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/AdvisoryCommitteeforPharmaceuticalScienceandClinicalPharmacology/ucm286697.htm.

34. Accessed December 31, 2012, at: www.nigms.nih.gov/nr/rdonlyres/. . ./systemspharmawpsorger2011.pdf.

35. Aberrnethy DR, Woodcock J, Lesko LJ. 2011 Pharmacologicalmechanism-based drug safety assessment and prediction. ClinPharmacol Ther 89:793–796.

36. Bai JP, Abernethy DR. 2013. Systems pharmacology to predictdrug toxicity: Integration across levels of biological organiza-tion. Ann Rev Pharmacol Toxicol 53:22.1–22.23.

37. Zhichkin PE, Athey BD, Avigan MI, Abernethy DR. 2012.Needs for an expanded ontology-based classification of adversedrug reactions and related mechanisms. Clin Pharmacol Ther91:963–965.

38. Sarntivijai S, Xiang Z, Shedden KA, Markel H, Omenn GS,Athey BD, He Y. 2012. Ontology-based combinatorial compar-ative analysis of adverse events associated with killed and liveinfluenza vaccines. PLoS ONE 7(11):e49941.

39. Accessed December 31, 2012, at: http://bioportal.bioontology.org.

40. Lounkine E, Keiser MJ, Whitebreat S, Mikhailov D, HamonJ, Jenkins JL, Lavan P, Weber E, Doad AK, Cote S, ShoichetBK, Urban L. 2012. Large-scale prediction and testing of drugactivity on side-effect targets. Nature 486(7403):361–367.

41. Armaiz-Pena GN, Allen JK, Cruz A, Stone RL, Nick AM,Lin YG, Han LY, Mangala LS, Villares GJ, Vivas-Mejia P,Rodriguez-Aguavo C, Nagaraia AS, Gharpure KM, Wu Z, En-glish RD, Soman KV, Shazhad MM, Zigler M, Deavers MT, ZienA, Soldatos TG, Jackson DB, Wirktorowicz JE, Torres-Lugo M,Young T, De Geest K, Gallick GE, Bar-Eli M, Lopez-BeresteinG, Cole SW, Lopez GE, Lutgendorf SK, Sood AK. 2013. Srcactivation by beta-adrenoreceptors is a key switch for tumourmetastasis. Nature Commun 4:1403.

42. Perakslis ED, Van Dam S, Szalma S. 2010. How informat-ics can potentiate precompetitive open-source collaboration tojump-start drug discovery and development. Clin PharmacolTher 87:614–616.

JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013 DOI 10.1002/jps

Page 12: Huang 2013

THE UTILITY OF MODELING AND SIMULATION IN DRUG DEVELOPMENT AND REGULATORY REVIEW 2923

43. Athey BD, Cavalcoli JD, Jagadish HV, Omenn GS, Mirel B,Kretzler M, Burant C, Isokpehi RD, DeLisi C. 2012. The NIHnational center for integrative biomedical informatics (NCIBI).J Am Med Inform Assoc 19:166–170.

44. Kligfield P, Green CL, Mortara J, Sager P, Stockbridge N,Li M, Zhang J, George S, Rodriguez I, Bloomfield D, KrucoffMW. 2010. The cardiac safety research consortium electrocar-diogram warehouse: Thorough QT database specifications andprinciples of use for algorithm development and testing. AmHeart J 160:1023–1028.

45. Force T, Kolaja KL. 2011. Cardiotoxicity of kinase inhibitors:The prediction and translation of preclinical models to clinicaloutcomes. Nat Rev Drug Discov 10:111–126.

46. Ghosh S, Matsuoka Y, Asai Y, Hsin KY, Kitano H. 2011. Soft-ware for systems biology: From tools to integrated platforms.Nat Rev Genet 12:821–832.

47. Accessed December 31, 2012. http://sbi.jp/aboutSBI.htm.48. Howell BA, Yang Y, Kumar R, Woodhead JL, Harrill AH,

Clewell HJ 3rd, Andersen ME, Siler SQ, Watkins PB. 2012.In vitro to in vivo extrapolation and species response compar-isons for drug-induced liver injury (DILI) using DILIsym: Amechanistic, mathematical model of DILI. J PharmacokinetPharmacodyn 39:527–541.

49. Kruhlak NL, Benz RD, Zhou H, Colatsky TJ. 2012. (Q)SARmodeling and safety assessment in regulatory review. ClinPharmacol Ther 91:529–534.

50. Quantitative and systems pharmacology at the post-genomeera; new approaches to discovering drugs and understandingtherapeutic mechanisms- an NIH white paper by the QSPworkshop group. October 2011. Accessed February 26, 2013,at: http://isp.hms.harvard.edu/wordpress/wp-content/uploads/2011/10/NIH-Systems-Pharma-Whitepaper-Sorger-et-al-2011.pdf.

51. US FDA CDER guidance for industry, population pharmacoki-netics. Accessed February 26, 2013, http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm064982.htm.

52. Zhao P, Rowland M, Huang SM. 2012. Best practice in the useof physiologically based pharmacokinetic modeling and simu-lation to address clinical pharmacology regulatory questions.Clin Pharmacol Ther 92:17–20.

53. CDER Guidance for industry. Drug interaction studies-study design, data analysis, implications for dosing, andlabeling recommendations. Accessed February 1, 2013,at: http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm064982.htm.

54. CDER Guidance for industry. Clinical pharmacogenomics:premarket evaluation in early-phase clinical studiesand recommendations for labeling. Accessed February1, 2013, at: http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm064982.htm.

55. US FDA CDER clinical pharmacology guidance page. AccessedFebruary 26, 2013, at: http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm064982.htm.

56. US FDA CDER guidance page: International conferenceon Harminisation- Efficacy. Accessed February 26, 2013,at: http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm065004.htm.

57. Woosley RL. 2012. Is it possible for FDA regulatory scientistsand industry scientists to work together? Clin Pharmacol Ther91:390–392.

58. Huang S-M, Woodcock J. 2010. Commentary on ITC mem-brane transporters in drug development; report from the FDAcritical path initiative sponsored workshop. Nat Rev Drug Dis-cov 9:175–176.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 102, NO. 9, SEPTEMBER 2013