+ All Categories
Home > Documents > Pooled Population Pharmacokinetic Analysis of Phase I, II and III Studies of Linifanib in Cancer...

Pooled Population Pharmacokinetic Analysis of Phase I, II and III Studies of Linifanib in Cancer...

Date post: 24-Jan-2017
Category:
Upload: dawn
View: 213 times
Download: 1 times
Share this document with a friend
13
ORIGINAL RESEARCH ARTICLE Pooled Population Pharmacokinetic Analysis of Phase I, II and III Studies of Linifanib in Cancer Patients Ahmed Hamed Salem Denise Koenig Dawn Carlson Published online: 4 December 2013 Ó Springer International Publishing Switzerland 2013 Abstract Background and Objective Linifanib is a multi-targeted receptor tyrosine kinase inhibitor, which can inhibit members of the vascular endothelial growth factor and platelet-derived growth factor receptor families. The objective of this analysis was to characterize the population pharmacokinetics of linifanib in cancer patients. Methods We pooled 7,351 linifanib plasma concentra- tions from 1,010 cancer patients enrolled in 13 clinical studies. Population pharmacokinetic modelling was per- formed using NONMEM version 7.2. The covariates that were screened included the cancer type, co-medications, creatinine clearance, formulation, fed status, liver function markers (bilirubin, blood urea nitrogen [BUN], aspartate aminotransferase [AST], alanine aminotransferase [ALT]), albumin, age, sex, race, body weight, surface area and body mass index. Results A two-compartment model with first-order absorption and disposition best described linifanib phar- macokinetics. An increase in body weight was associated with less than proportional increases in volumes of distri- bution. Subjects with hepatocellular carcinoma and renal cell carcinoma were estimated to have 63 and 86 % larger volumes of distribution, respectively, than subjects with the other cancer types. Females had 25 % slower oral clear- ance (CL/F) than males, while subjects with colorectal cancer had 41 % faster CL/F than other subjects. For linifanib bioavailability, subjects with refractory acute myeloid leukaemia or myelodysplastic syndrome had 43 % lower bioavailability, evening doses were associated with 27 % lower bioavailability than morning doses, and administration of linifanib under fed conditions decreased the bioavailability by 14 %. Finally, the oral solution for- mulation showed two-fold faster absorption than the tablet formulations. Conclusion The use of mixed-effects modelling allowed robust assessment of the impact of the concomitant effects of body size, different cancer types, formulation, diurnal variation, sex and food on linifanib pharmacokinetics. The developed population pharmacokinetic model describes linifanib concentrations adequately and can be used to conduct simulations or to evaluate the linifanib exposure– response relationship. 1 Introduction Angiogenesis is essential for tumour growth and metasta- sis. Lack of adequate vasculature results in tumours becoming necrotic or apoptotic, and restrict the tumour size [1, 2]. Angiogenesis involves an imbalance between anti- angiogenic and proangiogenic molecules such as vascular endothelial growth factor (VEGF) secreted from tumour cells [3]. In addition, platelet-derived growth factor (PDGF) stimulates tumour growth and enhances angio- genesis by facilitating pericyte coverage of new micro- vessels [4, 5]. Thus, inhibition of VEGF and PDGF receptors (VEGFR and PDGFR) has been a compelling target for cancer therapy [6, 7]. Results from clinical studies of bevacizumab, a selective monoclonal antibody against VEGFR, validated the A. H. Salem (&) D. Koenig D. Carlson Clinical Development, AbbVie Inc., 1 North Waukegan Road, AP13A-3, Dept. R4PK, North Chicago, IL 60064, USA e-mail: [email protected] A. H. Salem Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt Clin Pharmacokinet (2014) 53:347–359 DOI 10.1007/s40262-013-0121-2
Transcript

ORIGINAL RESEARCH ARTICLE

Pooled Population Pharmacokinetic Analysis of Phase I, II and IIIStudies of Linifanib in Cancer Patients

Ahmed Hamed Salem • Denise Koenig • Dawn Carlson

Published online: 4 December 2013

� Springer International Publishing Switzerland 2013

Abstract

Background and Objective Linifanib is a multi-targeted

receptor tyrosine kinase inhibitor, which can inhibit

members of the vascular endothelial growth factor and

platelet-derived growth factor receptor families. The

objective of this analysis was to characterize the population

pharmacokinetics of linifanib in cancer patients.

Methods We pooled 7,351 linifanib plasma concentra-

tions from 1,010 cancer patients enrolled in 13 clinical

studies. Population pharmacokinetic modelling was per-

formed using NONMEM version 7.2. The covariates that

were screened included the cancer type, co-medications,

creatinine clearance, formulation, fed status, liver function

markers (bilirubin, blood urea nitrogen [BUN], aspartate

aminotransferase [AST], alanine aminotransferase [ALT]),

albumin, age, sex, race, body weight, surface area and body

mass index.

Results A two-compartment model with first-order

absorption and disposition best described linifanib phar-

macokinetics. An increase in body weight was associated

with less than proportional increases in volumes of distri-

bution. Subjects with hepatocellular carcinoma and renal

cell carcinoma were estimated to have 63 and 86 % larger

volumes of distribution, respectively, than subjects with the

other cancer types. Females had 25 % slower oral clear-

ance (CL/F) than males, while subjects with colorectal

cancer had 41 % faster CL/F than other subjects. For

linifanib bioavailability, subjects with refractory acute

myeloid leukaemia or myelodysplastic syndrome had 43 %

lower bioavailability, evening doses were associated with

27 % lower bioavailability than morning doses, and

administration of linifanib under fed conditions decreased

the bioavailability by 14 %. Finally, the oral solution for-

mulation showed two-fold faster absorption than the tablet

formulations.

Conclusion The use of mixed-effects modelling allowed

robust assessment of the impact of the concomitant effects

of body size, different cancer types, formulation, diurnal

variation, sex and food on linifanib pharmacokinetics. The

developed population pharmacokinetic model describes

linifanib concentrations adequately and can be used to

conduct simulations or to evaluate the linifanib exposure–

response relationship.

1 Introduction

Angiogenesis is essential for tumour growth and metasta-

sis. Lack of adequate vasculature results in tumours

becoming necrotic or apoptotic, and restrict the tumour size

[1, 2]. Angiogenesis involves an imbalance between anti-

angiogenic and proangiogenic molecules such as vascular

endothelial growth factor (VEGF) secreted from tumour

cells [3]. In addition, platelet-derived growth factor

(PDGF) stimulates tumour growth and enhances angio-

genesis by facilitating pericyte coverage of new micro-

vessels [4, 5]. Thus, inhibition of VEGF and PDGF

receptors (VEGFR and PDGFR) has been a compelling

target for cancer therapy [6, 7].

Results from clinical studies of bevacizumab, a selective

monoclonal antibody against VEGFR, validated the

A. H. Salem (&) � D. Koenig � D. Carlson

Clinical Development, AbbVie Inc., 1 North Waukegan Road,

AP13A-3, Dept. R4PK, North Chicago, IL 60064, USA

e-mail: [email protected]

A. H. Salem

Department of Clinical Pharmacy, Faculty of Pharmacy,

Ain Shams University, Cairo, Egypt

Clin Pharmacokinet (2014) 53:347–359

DOI 10.1007/s40262-013-0121-2

targeting of VEGF-induced angiogenesis as an effective

anti-cancer therapeutic strategy [8–10]. Combined inhi-

bition of VEGFR and PDGFR is hypothesized to have a

greater antitumour effect than inhibition of individual

receptors [11]. In fact, there are three receptor tyrosine

kinase inhibitors—sorafenib, sunitinib and pazopanib—

that target both VEGFR and PDGFR, and they are already

approved for treatment of various solid tumour types,

with many more being in development. Linifanib (ABT-

869) is an orally active, multi-targeted receptor tyrosine

kinase inhibitor, which inhibits members of the VEGFR

and PDGFR families [12]. Linifanib exhibits more

selective inhibitory activity than other small molecules

targeting VEGFR and PDGFR tyrosine kinases, with less

activity against other unrelated tyrosine or serine/threo-

nine kinases [13]. Linifanib has demonstrated potent

antiproliferative and apoptotic effects on cancer cells and

has exhibited efficacy in human fibrosarcoma and breast,

colon and small cell lung carcinoma xenograft models

[13]. Evidence of encouraging clinical activity of linifanib

monotherapy has been demonstrated in relapsed or

refractory non-small cell lung cancer (NSCLC), hepato-

cellular carcinoma (HCC) and sunitinib-resistant renal cell

carcinoma (RCC) [14, 15].

Pharmacokinetic assessments in cancer patients have

shown that linifanib is rapidly absorbed, with an average

time to reach the peak concentration (tmax) of 2–3 h [16,

17]. The elimination half-life of linifanib ranged from 13.9

to 24 h in clinical studies [17, 18]. Linifanib pharmacoki-

netics showed dose proportionality over a 0.1–0.3 mg/kg

dose range [19]. Linifanib is predominantly metabolized,

with urinary recovery analysis showing that less than 5 %

of the linifanib dose is recovered in the urine as the

unchanged drug and metabolite [20]. The main systemic

metabolite for linifanib is the carboxylate metabolite [17].

The objectives of this analysis were to integrate the li-

nifanib concentration–time data from 13 clinical studies to

characterize the population pharmacokinetics of linifanib,

including identifying the structural pharmacokinetic model,

estimating pharmacokinetic parameters and associated

inter-individual variability, and testing patient demo-

graphics and covariates for their potential influence on

linifanib pharmacokinetics.

2 Methods

2.1 Clinical Studies and Patient Population

The population pharmacokinetic analysis included linifanib

plasma concentration data from 1,010 adults who partici-

pated in six phase I, six phase II and one phase III linifanib

clinical trials.

Table 1 summarizes the clinical studies used in this

analysis and their dosing and sampling schemes. The study

protocols were approved by the institutional review boards

of the individual study sites, and written informed consent

was obtained from each subject prior to enrolment. All

subjects were older than 18 years, with a histologically

confirmed malignancy.

2.2 Sample Collection and Quantification

Blood samples were collected, via venipuncture or a cen-

tral line, into ethylenediaminetetraacetic acid (EDTA)

tubes and stored on ice until centrifugation. Plasma sam-

ples were then stored at approximately -20 �C until ana-

lysis. Samples from all studies were analysed for plasma

concentrations of linifanib, using a validated liquid chro-

matography with tandem mass spectrometric detection

assay. The coefficient of variation was B7.7 %, indicating

the precision of the assay. The accuracy of the assay was

101.7 % at the lower limit of quantification (LLQ) and

ranged between 96.7 and 102.2 % at higher standard levels.

The LLQ was 1 ng/mL. Observations below the LLQ were

not included in the analysis.

2.3 Nonlinear Mixed-Effects Modelling

The population pharmacokinetic model was built using

nonlinear mixed-effects modelling in NONMEM ver-

sion 7.2 software (Icon Development Solutions, Ellicott

City, MD, USA). The first-order conditional estimation

method with interaction (FOCEI) was employed within

NONMEM. Diagnostic graphs and additional statistical

analyses were conducted using SAS version 9.3 and

R version 2.15.2 software. Development of the population

pharmacokinetic model started with construction of the

base model, including the structural pharmacokinetic

model and models for the inter-individual and residual

variabilities. Once the base model was developed, covari-

ate models were developed to explain the inter-individual

and residual variabilities.

2.3.1 Development of the Base Model

After the dose proportionality of linifanib was established,

standard linear compartmental models with first-order

absorption and elimination (ADVAN2, ADVAN4 or

ADVAN12) were evaluated for describing linifanib phar-

macokinetics. Different structures of the X matrix were

explored. Inter-individual variability in pharmacokinetic

parameters was modelled using an exponential error model

as shown for oral clearance (CL/F) in Eq. 1:

CL=F ¼ h1 � expðg1Þ ð1Þ

348 A. H. Salem et al.

Ta

ble

1C

har

acte

rist

ics

of

the

clin

ical

tria

lsin

clu

ded

inth

ean

aly

sis

Stu

dy

no

.S

ubje

cts

(n)

Stu

dy

po

pula

tio

nD

ose

sP

har

mac

ok

inet

icsa

mp

lin

gti

mes

Ag

e[y

ears

]aB

od

yw

eig

ht

[kg

]aS

ex[n

;m

ale/

fem

ale]

Rac

e[n

;W

hit

e/A

sian

/oth

er]

Ph

ase

Ist

ud

ies

13

3A

dv

ance

dn

on

-hae

mat

olo

gic

alm

alig

nan

cies

10

mg

;0

.1,

0.2

5,

0.3

mg

/kg

Inte

nsi

ve

(cycl

e1

day

1:

0,

0.5

,1

,2

,3

,4

,6

,8

,2

4h

;cy

cle

1d

ay1

5:

0,

0.5

,1

,2

,3

,4

,6

,8

h;

add

itio

nal

sam

ple

so

ncy

cle

3d

ay1

and

ever

y2nd

trea

tmen

tper

iod

ther

eaft

er)

57

.3(1

1.6

)

56

{2

9–7

6}

57

.7(1

4.1

)

56

{3

6–

10

7}

16

/17

0/3

1/2

24

4A

ML

or

MD

S1

0,

12

.5,

15

,2

0m

gIn

ten

siv

e(c

ycl

e1

day

1:

0,

0.2

5,

0.5

,1

,2

,3

,4

,6

,8

,2

4h

;cy

cle

1d

ay8

:0

,0

.25

,0

.5,

1,

2,

3,

4,

6,

8h

;ad

dit

ion

alsa

mp

les

on

cycl

e3

day

1an

dat

the

end

of

ever

y2

nd

cycl

eth

erea

fter

)

56

.4(1

5.7

)

59

{2

3–8

1}

78

.8(1

9.0

)

75

{4

6–

12

8}

29

/15

36

/7/1

31

8S

oli

dtu

mo

urs

0.0

5,

0.1

0,

0.2

0,

0.2

5m

g/k

gIn

ten

siv

e(c

ycl

e1

day

1:

0,

0.5

,1

,2

,3

,4

,6

,8

,2

4h

;cy

cle

1d

ay1

5:

0,

0.5

,1

,2

,3

,4

,6

,8

h)

52

.9(9

.99

)

52

{3

8–6

9}

57

.6(1

0.4

)

56

{4

5–

78

}

6/1

20

/18

/0

43

4A

dvan

ced

or

met

asta

tic

soli

dtu

mours

0.2

5m

g/k

gIn

tensi

ve

(day

1:

0.5

,1,

2,

3,

4,

6,

8,

10

,1

2,

24

,4

8,

72

h;

day

7:

0,

0.5

,1

,2

,3

,4

,6

,8

,1

0,

12

,2

4,

48

,7

2h

;d

ay2

1an

dd

ay3

0:

0,

1,

2,

3,

4,

6,

8h

)

60

.2(1

2.1

)

58

{3

6–8

1}

77

.5(2

0.5

)

71

{5

2–

14

7}

13

/21

31

/1/2

51

3A

dvan

ced

or

met

asta

tic

soli

dtu

mours

17.5

mg

Inte

nsi

ve

(day

1o

fea

chper

iod:

0,

0.5

,1

,2

,3

,4

,6

,8

,1

0,

12

,2

4,

48

,7

2,

96

h)

64

.6(8

.93

)

62

{4

9–7

8}

77

.5(1

7.8

)

77

{5

1–

10

7}

8/5

12

/1/0

61

0N

SC

LC

7.5

,1

2.5

mg

Inte

nsi

ve

(cy

cle

1d

ay2

1an

dcy

cle

2d

ay1

:0

,2

,3

,4

,8

,2

4h

)5

8.4

(9.0

3)

60

{4

3–6

9}

61

.7(1

2.9

)

62

{4

0–

79

}

7/3

0/1

0/0

Ph

ase

IIst

ud

ies

73

9H

CC

0.2

5m

g/k

gIn

ten

siv

ein

27

sub

ject

s(d

ay1

:0

,1

,2

,3

,4

,6

,8

,2

4,

48

h)

Sp

arse

in1

2su

bje

cts

(wee

ks

2an

d4

day

1,

end

of

wee

k8

)

58

.6(1

3.8

)

63

{2

0–8

1}

63

.0(1

3.9

)

59

{4

2–

11

5}

32

/72

/37

/0

81

28

NS

CL

C0

.10

,0

.25

mg

/kg

Sp

arse

(wee

ks

2,

3,

4,

5,

8)

61

.1(9

.92

)

62

{3

3–8

5}

71

.3(1

9.3

)

69

{3

5–

16

9}

75

/53

75

/47

/6

94

1R

CC

0.2

5m

g/k

gS

par

se(d

ays

8,

15

,2

9,

wee

ks

8an

d1

2)

59

.3(8

.54

)

60

{4

0–8

0}

88

.1(2

1.7

)

83

{5

7–

15

9}

34

/73

3/1

/7

10

10

Lo

call

yre

curr

ent

or

met

asta

tic

bre

ast

cance

r0

.15

,0

.20

mg

/kg

Inte

nsi

ve

(cy

cle

1d

ay2

8an

dcy

cle

2d

ay1

5:

0,

1,

2,

3,

4,

6,

8,

24

h)

53

.9(1

1.8

)

57

{3

0–7

3}

67

.2(1

7.6

)

64

{4

8–

10

7}

0/1

01

/2/7

11

10

6C

RC

7.5

,1

2.5

mg

Inte

nsi

ve

in8

sub

ject

s(c

ycl

e1

day

14

:0

,1

,2

,3

,4

,6

,8

,2

4h

;cy

cle

2d

ay1

:1

,2

,3

,4

,6

,8

,2

4h

)

Sp

arse

in9

8su

bje

cts

(cy

cle

1d

ay1

:0

.75

,2

,6

h;

cycl

es2

,3

,4

,6

,8

day

1:

0,

2h

;4

cycl

esth

erea

fter

day

1:

0h

)

58

.5(1

0.7

)

59

{3

1–8

1}

73

.0(1

4.0

)

72

{4

7–

10

6}

61

/45

79

/23

/4

PopPK Analysis of Linifanib in Cancer Patients 349

where h1 is the typical value (population mean) of CL/F and

g1 is an inter-individual random effect. The g values were

assumed to be independently, identically distributed, with

means of 0 and variances of x2: g * N(0, x2).

Residual variability was modelled using an additive

error model (Eq. 2), a proportional error model (constant

coefficient of variation, Eq. 3) or a combined additive and

proportional error model (Eq. 4), as follows:

Cij ¼ Cij þ eij ð2Þ

Cij ¼ Cij � ð1þ eijÞ ð3Þ

Cij ¼ Cij � ð1þ e1ijÞ þ e2ij ð4Þ

where Cij is the jth measured plasma concentration in indi-

vidual i, Cij is the jth model-predicted value in individual i, eij

is the residual random error for individual i and measurement

j, e1ij is the proportional component, and e2ij is the additive

component of the residual random error. The e values were

assumed to be independently and identically distributed, with

means of 0 and variances of r2: e * N(0, r2).

2.3.2 Identification of Significant Covariates

Empirical Bayesian estimates of individual parameters of

the base model were calculated by the posterior conditional

estimation technique (POSTHOC) in NONMEM, and their

association with pharmacokinetic parameters was investi-

gated. The covariates that were screened included the

cancer type, co-medications, creatinine clearance (CLCR),

linifanib formulation, fed status, liver function markers

(bilirubin, blood urea nitrogen [BUN], aspartate amino-

transferase [AST] and alanine aminotransferase [ALT]

levels), albumin level, age, sex, race, total body weight

(WT), body surface area and body mass index. Linifanib

exposures were previously reported to be higher following

morning administration than following evening adminis-

tration [21]. Therefore, we also tested the time of admin-

istration as a potential covariate for linifanib

bioavailability. Covariate modelling was performed using

the forward-inclusion, backward-elimination approach.

Power models were used for continuous covariates, with

the covariate scaled by the typical value, as shown in the

following example (Eq. 5):

TVVi ¼ h1 �WTi

70

� �h2

ð5Þ

where TVVi is the typical value of the apparent volume of

distribution (Vd/F) for an individual with WTi, and h1 is the

typical value of Vd/F for a 70 kg individual.

Dichotomous and categorical covariates were intro-

duced multiplicatively in the model via an indicator vari-

able. For example (Eq. 6):Ta

ble

1co

nti

nu

ed

Stu

dy

no

.S

ubje

cts

(n)

Stu

dy

po

pula

tio

nD

ose

sP

har

mac

ok

inet

icsa

mp

lin

gti

mes

Ag

e[y

ears

]aB

od

yw

eig

ht

[kg

]aS

ex[n

;m

ale/

fem

ale]

Rac

e[n

;W

hit

e/A

sian

/oth

er]

12

94

NS

CL

C7

.5,

12

.5m

gIn

ten

siv

ein

10

sub

ject

s(c

ycl

e1

day

21

and

cycl

e2

day

1:

0,

1,

2,

3,

4,

6,

8,

24

h)

Sp

arse

in8

4su

bje

cts

(cy

cle

1,

2,

3,

4d

ay1

:3

,6

h;

cycl

e1

day

s8

and

15

:0

h;

ever

ytw

ocy

cles

afte

rcy

cle

4d

ay1

)

59

.8(9

.23

)

60

{3

5–7

9}

69

.4(1

4.5

)

69

{4

1–

11

5}

55

/39

83

/6/5

Ph

ase

III

stud

y

13

44

0H

CC

17

.5m

gS

par

se(d

ay1

of

wee

k1

and

day

1o

fw

eek

3)

58

.3(1

1.7

)

59

{2

1–8

4}

68

.1(1

3.2

)

65

{3

8–

12

3}

38

0/6

01

29

/305

/6

AM

Lac

ute

myel

ogen

ous

leukae

mia

,C

RC

colo

rect

alca

nce

r,H

CC

hep

ato

cell

ula

rca

rcin

om

a,M

DS

myel

od

ysp

last

icsy

ndro

me,

NSC

LC

non-s

mal

lce

lllu

ng

cance

r,R

CC

renal

cell

carc

ino

ma,

SD

stan

dar

dd

evia

tio

na

Val

ues

are

exp

ress

edas

mea

n(S

D);

med

ian

{ra

ng

e}

350 A. H. Salem et al.

TVCLi ¼ h1

IF CAT:EQ:1ð ÞTVCLi ¼ h1 � h2

IF CAT:EQ:2ð ÞTVCLi ¼ h1 � h3

ð6Þ

where TVCLi is the typical value of clearance for indi-

vidual i, h1 is the typical value of CL/F for the reference

group with categorical covariate (CAT) = 0, and h2 and h3

represent the fixed effects of the categorical covariate.

2.3.3 Model Selection and Evaluation of the Final Model

Model selection was based on evaluation of the goodness-

of-fit plots, and attainment of physiologically reasonable

and statistically significant parameter estimates. In addi-

tion, the differences in the objective function value (OFV)

were used to guide model building. Since the OFV pro-

vided by NONMEM is approximately v2 distributed, the

likelihood ratio test was used for hypothesis testing to

discriminate among alternative nested population pharma-

cokinetic models. Likelihood ratio tests were assessed at

the 0.01 significance level (OFV drop [6.63 for a change

of one degree of freedom), but tests in the backward-

elimination step of the covariate selection procedure were

assessed at the 0.001 significance level (OFV increase of

10.83 for a change of one degree of freedom). Comparison

between non-hierarchical models was performed using the

Akaike information criterion.

The robustness of the final model parameter estimates

was evaluated using bootstrapping. A total of 1,000 boot-

strap replicates were constructed by randomly sampling

(with replacement) 1,010 subjects from the original dataset.

Model parameters were estimated for each bootstrap rep-

licate, and the resulting values were used to calculate

medians and confidence intervals. Bootstrap statistics were

Table 2 Patient demographics and clinical characteristics

Patient characteristic n (%) Mean (SD) Median Range

Age (years) 1,010 58.8 (11.4) 60.0 20.0–85.0

Height (cm) 1,010 166.9 (8.9) 168.0 130.0–196.0

Body weight (kg) 1,010 70.0 (16.4) 67.0 35.0–169.0

Body mass index (kg/m2) 1,010 25.1 (5.1) 24.2 13.7–50.5

Sex

Female 294 (29.1)

Male 716 (70.9)

Race

Asian 489 (48.4)

Other 40 (4.0)

White 481 (47.6)

Formulation

Oral solution 258 (25.5)

Uncoated tablets 330 (32.7)

Coated tablets 453 (44.9)

Cancer type

Breast cancer 19 (1.9)

CRC 118 (11.7)

HCC 483 (47.8)

NSCLC 232 (23.0)

Solid tumours 68 (6.7)

AML/MDS 44 (4.4)

RCC 46 (4.6)

Bilirubin (mg/dL) 1,010 0.7 (0.47) 0.59 0.1–3.5

Albumin (g/dL) 1,010 4.0 (0.47) 4.00 2.1–5.1

Creatinine (mg/dL) 1,010 0.9 (0.24) 0.81 0.4–2.0

Aspartate aminotransferase (IU/L) 1,010 46.6 (35.3) 34.0 8.0–211.0

Alanine aminotransferase (IU/L) 1,010 36.0 (29.8) 27.0 5.0–221.0

Creatinine clearance (mL/min) 1,010 92.0 (31.3) 87.8 21.3–290.4

AML acute myelogenous leukaemia, CRC colorectal cancer, HCC hepatocellular carcinoma, MDS myelodysplastic syndrome, NSCLC non-small

cell lung cancer, RCC renal cell carcinoma, SD standard deviation

PopPK Analysis of Linifanib in Cancer Patients 351

based only on replicates that converged successfully.

Model parameters based on the original dataset were

compared against the bootstrap results.

The predictive performance of the final models and its

usefulness for describing observations was assessed using

prediction and variance-corrected visual predictive checks

(VPCs), where the final parameter estimates were used to

simulate 1,000 replicates of the observed dataset. Both

observations and the simulated data were normalized on

the typical model prediction for the median independent

variable in each bin in order to account for variation in

sampling times and predictive covariates introduced by

binning of the observations [22]. The median and the 5th

and 95th percentile concentrations of the simulated datasets

were then plotted against the original observations.

3 Results

3.1 Base Model

A total of 7,351 plasma concentrations from 1,010 cancer

patients were included in the analysis. The demographic

and clinical characteristics of the patient population

included in the analysis are summarized in Table 2.

A two-compartment disposition model with first-order

absorption and elimination best described the data. The

model was parameterized in terms of the absorption rate

constant (ka), apparent clearance from the central com-

partment (CL/F), the apparent volume of the central

compartment (Vc/F), apparent inter-compartmental clear-

ance (Q/F), and the apparent volume of the peripheral

compartment (Vp/F). The model provided a better fit than a

one-compartment model with an OFV drop of 160. Adding

a third compartment did not further improve the fit. Using a

lag time for describing a potential absorption delay led to

unstable parameter estimates, although it decreased the

OFV significantly. Therefore, it was decided not to include

the lag time in the model.

The residual unexplained variability was best charac-

terized using a combined additive and proportional error

model, while additive and proportional residual error

models provided inferior fits. The data supported including

inter-individual variability terms for CL/F, Vc/F and ka,

which were estimated with high precision. Estimating

inter-individual variability in Vp/F and Q/F was associated

Table 3 Final estimates of the population pharmacokinetic parameters obtained using NONMEM and bootstrap analysis of the final model

Parameter Original data result Bootstrap resulta

Estimate (%RSE) Median 95 % CI

CL/F (L/h) 2.82 (2.30) 2.82 2.76–2.87

Vc/F (L) 50.75 (4.40) 50.76 50.00–51.55

ka (L/h) 0.46 (8.70) 0.46 0.44–0.48

Q/F (L/h) 1.14 (30.00) 1.14 0.97–1.35

Vp/F (L) 10.36 (16.00) 10.38 9.34–11.34

Oral solution on ka 1.97 (14.00) 1.98 1.85–2.16

Time of dose administration on F 0.73 (1.70) 0.73 0.71–0.75

CRC on CL/F 1.41 (4.50) 1.41 1.37–1.45

Body weight on Vc/F and Vp/F 0.52 (18.00) 0.52 0.47–0.56

Sex on CL/F 0.75 (3.30) 0.75 0.74–0.76

HCC on Vc/F and Vp/F 1.63 (6.40) 1.63 1.60–1.65

Food condition on F 0.86 (1.80) 0.86 0.82–0.91

AML/MDS cancer on F 0.57 (5.20) 0.57 0.55–0.58

RCC on Vc/F and Vp/F 1.86 (13.00) 1.87 1.81–1.93

IIV on CL/F (%CV) 42 (5.30) 42 41–42

IIV on Vc/F (%CV) 39 (16.00) 39 37–40

IIV on ka (%CV) 97 (11.00) 97 94–99

Proportional component (%CV) 34 (0.80) 34 34–34

Additive component (ng/mL) 8 (7.70) 8 7–9

AML acute myelogenous leukaemia, CI confidence interval, CL/F apparent clearance from the central compartment, CRC colorectal cancer, CV

coefficient of variation, F bioavailability, HCC hepatocellular carcinoma, IIV inter-individual variability, ka absorption rate constant, MDS

myelodysplastic syndrome, NSCLC non-small cell lung cancer, Q/F apparent inter-compartmental clearance, RCC renal cell carcinoma, RSE

relative standard error, Vc/F apparent volume of the central compartment, Vp/F apparent volume of the peripheral compartmenta Based on 854/1,000 successful runs

352 A. H. Salem et al.

with unphysiological estimates of Vp/F and Q/F and hence

were not included in the model. Estimating the full

covariance matrix between CL/F, Vc/F and ka was

attempted; however, the covariance parameters were esti-

mated with poor precision. Therefore, covariance between

only CL/F and Vc/F was included throughout model

development, with their correlation estimated to be 0.52 in

the base model.

3.2 Covariate Model

Covariates identified by the univariate stepwise forward-

inclusion procedure as significant determinants of linifanib

pharmacokinetic parameters were as follows: sex, cancer

type (colorectal cancer [CRC]) and cytarabine co-admin-

istration as covariates on CL/F, WT, cancer type (HCC and

RCC) as covariates on Vc/F and Vp/F, formulation effect on

ka and cancer type (refractory acute myeloid leukaemia

[AML] or myelodysplastic syndrome [MDS]), time of dose

administration and fed status on bioavailability. No other

evaluated covariates were found to significantly improve

the goodness of fit. Removal of the cytarabine effect on

CL/F in the stepwise backward elimination was associated

with a nonsignificant increase in OFV (p value [0.05);

hence, the cytarabine effect was removed from the model.

After reaching the final model, we tested the need for

estimating the exponent of the allometric model for WT on

Vc/F and Vp/F to ensure model parsimony. Fixing the

exponents to 1, as suggested by some researchers, resulted

in a significant increase in the OFV (p value \0.001);

hence, estimating the exponent was deemed appropriate.

3.3 Final Model

The final model parameter estimates and the precision

associated with their estimation are shown in Table 3. Both

fixed and random effects were precisely estimated, with a

relative standard error (RSE) of 30 % or less. In the final

model, an increase in WT was associated with a less than

proportional increase in Vc/F and Vp/F. Subjects with HCC

and RCC were estimated to have 63 and 86 % larger vol-

umes of distribution, respectively, than subjects with the

other cancer types. With regard to CL/F, females had 25 %

slower clearance than males. Furthermore, subjects with

colorectal cancer had 41 % faster CL/F than non-colorectal

cancer subjects.

For linifanib bioavailability, subjects with relapsed/

refractory AML/MDS had 43 % lower bioavailability, and

evening doses were associated with 27 % lower bioavail-

ability than morning doses, while administration of linifa-

nib under fed conditions decreased the bioavailability by

14 %. Finally, the oral solution formulation showed

approximately two-fold faster absorption than the tablet

formulations.

Relative to the base model, the covariates explained

27, 9 and 29 % of the variability of Vc/F, CL/F and the

correlation between Vc/F, CL/F, respectively. The

effects of some covariates on linifanib CL/F and

Vc/F are shown in Fig. 1. Figure 2 demonstrates the

effect of sex on CL/F and the lack of association

between body weight and CL/F.

The final equations for the typical values (TVs) of the

structural model parameters are presented in Eqs. 7, 8, 9

and 10:

TVCL=F ¼ 2:82� hCRC � hSEX ð7Þ

where:

hCRC ¼1:41; colorectal cancer

1; else

�;

hSEX ¼0:75; female

1; male

TVVc=F ¼ 50:75� WT=70ð Þ0:52�hHRC ð8Þ

where:

hHRC ¼1:63;1:86;

1;

hepatocellular carcinoma

renal cell carcinoma

else

8<:

TVka ¼ 0:46� hSOL ð9Þ

where:

hSOL ¼1:97; oral solution

1; tablets

TVF ¼ hAMPM � hFCOND � hAML MDS ð10Þ

where:

hAMPM ¼0:73; evening dose

1; morning dose

hFCOND ¼0:86; non-fasting

1; else

hAML MDS ¼0:57; Refractory AML/MDS cancer

1; else

The goodness of fit for the final model was evaluated

graphically (Figs. 3, 4). The individual and population

predicted linifanib concentrations versus the observed

concentrations were randomly distributed across the line

of unity, indicating that the model adequately described the

observations over the entire linifanib concentration range.

The conditional weighted residuals plots showed

symmetrical distribution and no time- or concentration-

related trends.

PopPK Analysis of Linifanib in Cancer Patients 353

Fig. 1 Effects of cancer type,

body weight and sex on

linifanib pharmacokinetic

parameters. The boxes represent

the 25th, 50th and 75th

percentiles; the whiskers

represent the lowest datum still

within 1.5 interquartile ranges

(IQRs) of the lower quartile, and

the highest datum still within

1.5 IQRs of the upper quartile

range; the bullets represents

outliers. AML acute

myelogenous leukaemia, CL/F

apparent clearance from the

central compartment, CRC

colorectal cancer, HCC

hepatocellular carcinoma, MDS

myelodysplastic syndrome,

NSCLC non-small cell lung

cancer, RCC renal cell

carcinoma, Vc/F apparent

volume of the central

compartment

Fig. 2 Relationships between

sex and body weight versus

ETA on apparent clearance

from the central compartment

(CL/F) in the base and final

models. The boxes represent the

25th, 50th and 75th percentiles;

the whiskers represent the

lowest datum still within 1.5

interquartile ranges (IQRs) of

the lower quartile and the

highest datum still within 1.5

IQRs of the upper quartile

range; the bullets represents

outliers

354 A. H. Salem et al.

Fig. 3 Diagnostic plots of the

final model

Fig. 4 Population predicted

linifanib concentration–time

after last dose profiles for all

studies. The solid lines ? error

bars denote the medians and

90 % percentiles of the

population predicted linifanib

concentrations, and the circles

denote the observed linifanib

concentrations

PopPK Analysis of Linifanib in Cancer Patients 355

In order to confirm the stability of the model preci-

sion of the estimated pharmacokinetic parameters, a

non-parametric bootstrap analysis was performed, and

85 % of the bootstrap replicates converged successfully.

In accordance with the estimated standard error of

estimate (SEE) values for pharmacokinetic parameters in

the linifanib pharmacokinetic model, the bootstrap

showed narrow confidence intervals for all parameters.

The median and the 5th and 95th percentiles of the

parameter estimates from the fit of the final model to the

bootstrap samples are shown in Table 3. The asymptotic

estimates obtained from the original dataset showed

close agreement with the median and were all included

within the 5th to the 95th percentiles of the bootstrap-

ping values, indicating model stability. None of the

95 % confidence intervals for the parameters from the

bootstrap datasets included zero, confirming the robust-

ness of the parameters.

In the VPC plots shown in Fig. 5, the 5th, 50th and 95th

percentiles of the prediction-corrected observations were in

close agreement with the confidence intervals of the 5th,

50th and 95th percentiles of the prediction-corrected sim-

ulated data, indicating a robust predictive ability of the

model to describe linifanib concentrations. Only 4 % of the

data were below the 5th percentile, and 5 % of the data

were above the 95th percentile of the predictions.

4 Discussion

Using non-linear mixed-effects analysis, linifanib concen-

tration–time data from 13 clinical studies were integrated

across the different development phases in order to char-

acterize the effect of covariates on linifanib pharmacoki-

netics. The developed model showed high parameter

estimate precision, as well as good predictability and hence

will be of immense value in predicting linifanib pharma-

cokinetics in various cancer patient populations.

The final structural pharmacokinetic model was a two-

compartment model with first-order absorption and elimi-

nation. Compared with the one-compartment model, the

use of a two-compartment model in our analysis was

associated with a better fit to individual profiles and a

larger reduction in the objective function value. A previous

analysis based on a phase II study used a one-compartment

model for linifanib in subjects with NSCLC [23]. The

sparse data included in the previous analysis may not have

allowed characterization of the biphasic disposition of

linifanib.

The population estimate of linifanib CL/F in the final

model was 2.8 L/h. Previous pharmacokinetic assessments

of individual studies have shown similar estimates. In a

phase I study in refractory solid tumours, linifanib

CL/F was 2.7 (±1.2) L/h [17]. In phase II studies, linifanib

Fig. 5 Prediction and variance-

corrected visual predictive

check for the final model. The

circles denote the prediction-

corrected observations; the lines

denote the 5th, 50th, and 95th

percentiles of the prediction-

corrected observed data; the

shaded areas denote the

confidence intervals of the 5th,

50th, and 95th percentiles of the

prediction-corrected simulated

data

356 A. H. Salem et al.

CL/F ranged from 3 to 3.9 L/h [23, 24]. The steady-state

volume of distribution (Vss/F) of linifanib was estimated to

be 61.1 L. This may indicate extensive distribution of

linifanib in the body, which is consistent with its high

lipophilicity (Log D of 4.2 at pH of 7.4) [12]. The beta-

phase half-life of 17.2 h is supportive of the once-daily

dosing used throughout development.

We explored the effect of body size measures on

linifanib pharmacokinetic parameters. No association was

found between the body size measures and linifanib CL/F,

despite the wide body weight range of the subjects included

in the analysis (Table 1). This corroborates the switch of

linifanib dosing from body weight-guided dosing in phase I

and II studies to fixed dosing in the phase III study. On the

other hand, Vc/F and Vp/F were associated with body

weight, and the data supported the estimation of the

exponent of the allometric size model. Subjects with WT

that is 10 and 20 % larger than the population typical body

weight of 70 kg are expected to have 5 and 10 % larger

Vss/F values than the population typical estimate.

The final model also estimated that morning dosing was

associated with 27 % greater exposure than evening dos-

ing. This diurnal variation has been demonstrated previ-

ously in a phase I study, which assessed the impact of the

dose time on linifanib pharmacokinetics [21]. In order to

minimize variability and improve the tolerability of

linifanib, the hepatocellular carcinoma subjects enrolled in

the phase III study were instructed to administer the doses

in the evening.

Diurnal variation in pharmacokinetics, often referred to

as chronopharmacokinetics, is exhibited by several orally

administered anti-cancer drugs; such as 6-mercaptopurine,

busulfan and tegafur/uracil [25–29]. Moreover, some

anticancer drugs, such as fluorouracil and doxorubicin, also

show diurnal variation in their plasma levels although they

are administered by constant continuous intravenous infu-

sion [30–33]. It is hypothesized that such diurnal variations

are often masked by the high inter-individual variability

observed in cancer patients [34]. Diurnal variation has been

also reported with drugs from other classes [35, 36].

The lower linifanib exposure in the evening could be

explained by the reduction in the gastric emptying rate in

the evening, due to reduced enterokinesis [37]. The slower

gastric emptying rate in the evening was deemed respon-

sible for lower peak concentrations and longer tmax values

for many lipophilic drugs [37]. Another possible mecha-

nism is the circadian rhythm in the activity of metabolizing

enzymes. Animal studies have shown several-fold greater

activity in some microsomal oxidases during the dark span

than in the light span [38–40]. Previous population phar-

macokinetic analyses reported strong associations between

microsomal oxidases activity and the clearance of another

anticancer drug, gefitinib, with 60 % of the variability of

unbound gefitinib plasma concentrations being explained

by individual cytochrome P450 3A4 activity [41, 42].

This population analysis showed that the cancer type

was an important determinant of linifanib pharmacokinet-

ics. According to the final model, subjects with colorectal

cancer have 41 % faster linifanib oral clearance. Colorectal

cancer subjects included in the analysis were co-medicated

with mFOLFOX6 (oxaliplatin [85 mg/m2], folinic acid

[400 mg/m2] and 5 fluorouracil [400 mg/m2 IV bolus]) on

the first day of each cycle. In a lead-in cohort in this

population, linifanib pharmacokinetics was evaluated with

and without mFOLFOX6. Therefore, we explored the

possibility of a drug interaction between linifanib and

mFOLFOX6 being responsible for the increase in CL/F in

this population. However, mFOLFOX6 showed no effect

on linifanib pharmacokinetics. Furthermore, our analysis

demonstrated 43 % lesser bioavailability in subjects with

relapsed or refractory AML or MDS than in those with

other cancer types. The lesser bioavailability estimated in

this patient population is consistent with the reportedly

greater CL/F estimates in this population (4.1–6.9 L/h)

[20] than in patients with other cancer types (2.7–3.9 L/h)

[17, 23, 24]. We initially attempted to include the relapsed/

refractory AML/MDS cancer type as a covariate on CL/F;

however, including it on bioavailability provided a much

larger OFV reduction. The formulation and assay were

precluded as sources of the lesser bioavailability in this

population, since the same formulation and assay were

used in studies conducted in other populations. We

hypothesize that this reduction in oral bioavailability is due

to cytotoxic therapy-induced malabsorption. The subjects

with relapsed/refractory AML or MDS who were included

in our analysis had been heavily pretreated with cytotoxic

therapy. Cytotoxic therapy affects the dividing cells of the

gastrointestinal mucosa and has been reported to cause

intestinal epithelial damage in AML subjects [43, 44].

Intestinal injury induced by cytotoxic therapy has been

linked to reduced absorption and lesser bioavailability of

several agents, such as acyclovir, ciprofloxacin and

D-xylose [45–47].

Another difference in pharmacokinetics between cancer

types was the higher Vss/F value in subjects with HCC and

RCC. This could be related to fluid retention and increased

fluid volume during hepatic and renal impairment. In

addition, linifanib is highly protein bound ([99 %), and

drugs that are strongly bound to plasma constituents (e.g.

phenytoin, diazepam) are known to demonstrate an

increased volume of distribution in patients with liver or

kidney disease, which is due to lower plasma binding [48].

In order to investigate whether the increase in linifanib

Vd/F in subjects with HCC and RCC could be explained by

change in protein binding in these populations, we explored

the use of albumin concentrations as a covariate in the

PopPK Analysis of Linifanib in Cancer Patients 357

model. However, no relationship was shown between

albumin concentrations and linifanib Vc/F or CL/F. In

addition, plasma protein binding data from 13 subjects with

HCC (eight Child–Pugh class A and five Child–Pugh

class B) showed that [99.7 % of linifanib was protein

bound in plasma, which was similar to values in subjects

with normal hepatic function [49]. Therefore, we do not

believe that the larger Vd/F in HCC and RCC subjects is

linked to a decrease in protein binding.

An oral solution formulation was used initially during

linifanib clinical development, before tablet formulations

were developed. In alignment with the results of compar-

ative bioavailability studies, our population analysis

showed no difference in linifanib bioavailability among the

formulations. Nevertheless, the analysis showed a differ-

ence in the absorption rate constant between the formula-

tions, with the liquid formulation having two-fold faster

absorption than the tablet formulation. We attempted to

estimate different inter-individual variability for ka of the

oral solution compared with the tablet formulations; how-

ever, a high shrinkage ([70 %) in the inter-individual

variability of ka of the oral solution was observed. A model

with only one random effect for ka for all formulations

decreased the shrinkage, and hence the same inter-indi-

vidual variability was estimated for ka regardless of the

formulation.

Our analysis showed a sex effect on linifanib CL/F, with

males having 25 % faster CL/F than females. This differ-

ence is similar to that observed with another tyrosine

kinase inhibitor, erlotinib, where female subjects achieved

greater exposure (25–43 %) than male subjects [50]. Faster

clearance in male subjects for some drugs is often sec-

ondary to the body weight influence on clearance. In an

attempt to investigate whether the sex effect seen on

linifanib CL/F is due to a body weight effect, we explored

the relationship between linifanib CL/F and body weight.

However, no association was found between CL/F and

body weight (Fig. 2). Numerous clinical pharmacokinetic

studies have demonstrated sex differences in drug absorp-

tion and bioavailability for certain drugs [51]. Neverthe-

less, we do not believe that this is the underlying

mechanism of the sex effect on linifanib CL/F, since there

was no difference in Vd/F between males and females. We

hypothesize that sex-based differences in drug metabolism

play a role in the faster clearance observed in males.

In several clinical studies, linifanib was co-administered

with other anti-cancer agents, such as cytarabine, paclit-

axel, carboplatin and mFolfox6. Our analysis suggests that

these drugs do not affect linifanib pharmacokinetics. This

may be because of the multiple pathways involved in

linifanib metabolism. In addition, we found no difference

in pharmacokinetic parameters between Asians, who were

well represented in our dataset, and non-Asian populations.

Finally, there was no association between linifanib

CL/F and CLCR. This was anticipated, since urinary elimina-

tion is a minor pathway in linifanib pharmacokinetics, with

less than 5 % of the linifanib dose being recovered in urine

as the unchanged drug and metabolite [20].

5 Conclusion

The use of mixed-effects modelling allowed robust

assessment of the impact of the concomitant effects of

body size, different cancer types, formulation, diurnal

variation, sex and food. The developed population phar-

macokinetic model describes linifanib concentrations ade-

quately and can be used to conduct simulations or to

evaluate the linifanib exposure–response relationship.

Conflicts of Interest This study was sponsored by AbbVie Inc.

AbbVie Inc. contributed to the study design; research; data interpre-

tation; and writing, review and approval of the manuscript for pub-

lication. Ahmed Hamed Salem, Denise Koenig and Dawn Carlson

are employees of AbbVie Inc.

References

1. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases.

Nature. 2000;407(6801):249–57.

2. Zetter P, Bruce R. Angiogenesis and tumor metastasis. Annu Rev

Med. 1998;49(1):407–24.

3. Dvorak HF. Vascular permeability factor/vascular endothelial

growth factor: a critical cytokine in tumor angiogenesis and a

potential target for diagnosis and therapy. J Clin Oncol.

2002;20(21):4368–80.

4. Benjamin LE, Golijanin D, Itin A, Pode D, Keshet E. Selective

ablation of immature blood vessels in established human tumors

follows vascular endothelial growth factor withdrawal. J Clin

Invest. 1999;103(2):159.

5. Carmeliet P. Angiogenesis in health and disease. Nat Med.

2003;9(6):653–60.

6. Perona R. Cell signalling: growth factors and tyrosine kinase

receptors. Clin Transl Oncol. 2006;8(2):77–82.

7. Homsi J, Daud AI. Spectrum of activity and mechanism of action

of VEGF/PDGF inhibitors. Cancer Control. 2007;14(3):285.

8. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus

irinotecan, fluorouracil, and leucovorin for metastatic colorectal

cancer. N Engl J Med. 2004;350(23):2335–42.

9. Reck M, Von Pawel J, Zatloukal Pv, et al. Overall survival with

cisplatin–gemcitabine and bevacizumab or placebo as first-line

therapy for nonsquamous non-small-cell lung cancer: results from

a randomised phase III trial (AVAiL). Ann Oncol. 2010;21(9):

1804–9.

10. Sandler A, Gray R, Perry MC, et al. Paclitaxel–carboplatin alone

or with bevacizumab for non–small-cell lung cancer. N Engl J

Med. 2006;355(24):2542–50.

11. Erber R, Thurnher A, Katsen AD, et al. Combined inhibition of

VEGF and PDGF signaling enforces tumor vessel regression by

interfering with pericyte-mediated endothelial cell survival

mechanisms. FASEB J. 2004;18(2):338–40.

12. Dai Y, Hartandi K, Ji Z, et al. Discovery of N-(4-(3-amino-1

H-indazol-4-yl) phenyl)-N0-(2-fluoro-5-methylphenyl) urea

358 A. H. Salem et al.

(ABT-869), a 3-aminoindazole-based orally active multitargeted

receptor tyrosine kinase inhibitor. J Med Chem. 2007;50(7):

1584–97.

13. Albert DH, Tapang P, Magoc TJ, et al. Preclinical activity of

ABT-869, a multitargeted receptor tyrosine kinase inhibitor. Mol

Cancer Ther. 2006;5(4):995–1006.

14. Linifanib. Drugs R D. 2010;10(2):111–22.

15. Toh HC, Chen PJ, Carr BI, et al. Phase 2 trial of linifanib (ABT-

869) in patients with unresectable or metastatic hepatocellular

carcinoma. Cancer. 2013;119(2):380–7.

16. Asahina H, Tamura Y, Nokihara H, et al. An open-label, phase 1

study evaluating safety, tolerability, and pharmacokinetics of

linifanib (ABT-869) in Japanese patients with solid tumors.

Cancer Chemother Pharmacol. 2012;69(6):1477–86.

17. Wong CI, Koh TS, Soo R, et al. Phase I and biomarker study of

ABT-869, a multiple receptor tyrosine kinase inhibitor, in

patients with refractory solid malignancies. J Clin Oncol.

2009;27(28):4718–26.

18. Steinberg J, Tan E, Wei-Peng Y. Preliminary analysis of ABT-

869 safety, pharmacokinetics and efficacy in three phase 2 solid

tumor studies [abstract no. 477P]. 33rd Congress of the European

Society for Medical Oncology; Stockholm; 12–16 Sep 2008.

19. Humerickhouse R, Gupta N, Goh B. Phase 1 comparison of

pharmacokientics, safety and efficacy with low versus high doses

of ABT-869 in refractory solid tumors [abstract no. 476P]. 33rd

Congress of the European Society for Medical Oncology;

Stockholm; 12–16 Sep 2008.

20. Wang ES, Yee K, Koh LP, et al. Phase 1 trial of linifanib (ABT-

869) in patients with refractory or relapsed acute myeloid leu-

kemia. Leuk Lymphoma. 2012;53(8):1543–51.

21. Gupta N, Yan Z, LoRusso P, Ricker J, Carlson D, Pradhan R.

Assessment of the effect of food on the oral bioavailability and

assessment of diurnal variation in the pharmacokinetics of

linifanib. Mol Cancer Ther. 2009;8(12 Suppl):B53.

22. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction-

corrected visual predictive checks for diagnosing nonlinear

mixed-effects models. AAPS J. 2011;13(2):143–51.

23. Tan EH, Goss GD, Salgia R, et al. Phase 2 trial of linifanib

(ABT-869) in patients with advanced non-small cell lung cancer.

J Thorac Oncol. 2011;6(8):1418–25.

24. Tannir NM, Wong YN, Kollmannsberger CK, et al. Phase 2 trial

of linifanib (ABT-869) in patients with advanced renal cell cancer

after sunitinib failure. Eur J Cancer. 2011;47(18):2706–14.

25. Etienne-Grimaldi M, Cardot J, Francois E, et al. Chronophar-

macokinetics of oral tegafur and uracil in colorectal cancer

patients. Clin Pharmacol Ther. 2007;83(3):413–5.

26. Hassan M, Oberg G, Bekassy A, et al. Pharmacokinetics of high-

dose busulphan in relation to age and chronopharmacology.

Cancer Chemother Pharmacol. 1991;28(2):130–4.

27. Koren G, Langevin AM, Olivieri N, Giesbrecht E, Zipursky A,

Greenberg M. Diurnal variation in the pharmacokinetics and

myelotoxicity of mercaptopurine in children with acute lym-

phocytic leukemia. Arch Pediatr Adolesc Med. 1990;144(10):

1135.

28. Muggia FM, Wu X, Spicer D, et al. Phase I and pharmacokinetic

study of oral UFT, a combination of the 5-fluorouracil prodrug

tegafur and uracil. Clin Cancer Res. 1996;2(9):1461–7.

29. Vassal G, Challine D, Koscielny S, et al. Chronopharmacology of

high-dose busulfan in children. Cancer Res. 1993;53(7):1534–7.

30. Metzger G, Massari C, Etienne M-C, et al. Spontaneous or

imposed circadian changes in plasma concentrations of 5-fluo-

rouracil coadministered with folinic acid and oxaliplatin: rela-

tionship with mucosal toxicity in patients with cancer. Clin

Pharmacol Ther. 1994;56(2):190–201.

31. Petit E, Milano G, Levi F, Thyss A, Bailleul F, Schneider M.

Circadian rhythm-varying plasma concentration of 5-fluorouracil

during a five-day continuous venous infusion at a constant rate in

cancer patients. Cancer Res. 1988;48(6):1676–9.

32. Squalli A, Oustrin J, Houin G. Clinical chronopharmacokinetics of

doxorubicin (DXR). Annu Rev Chronopharmacol. 1989;5:393–6.

33. Levi F. Circadian rhythms in 5-fluorouracil pharmacology and

therapeutic applications. In: Rustum YM. Fluoropyrimidines in

cancer therapy. Totowa: Humana Press, 2003. p. 107–28.

34. Levi F, Okyar A, Dulong S, Innominato PF, Clairambault J.

Circadian timing in cancer treatments. Annu Rev Pharmacol

Toxicol. 2010;50:377–421.

35. Zhang C, Denti P, Decloedt E, et al. Model-based approach to

dose optimization of lopinavir/ritonavir when co-administered

with rifampicin. Br J Clin Pharmacol. 2012;73(5):758–67.

36. Iwahori T, Takeuchi H, Matsuno N, et al. Pharmacokinetic dif-

ferences between morning and evening administration of cyclo-

sporine and tacrolimus therapy. Transplant Proc. 2005;37:

1739–40.

37. Lemmer B. Chronopharmacokinetics: implications for drug

treatment. J Pharm Pharmacol. 1999;51(8):887–90.

38. Belanger P. Chronobiological variation in the hepatic elimination

of drugs and toxic chemical agents. Annu Rev Chronopharmacol.

1988;4:1–46.

39. Radzialowski FM, Bousquet WF. Daily rhythmic variation in

hepatic drug metabolism in the rat and mouse. J Pharmacol Exp

Ther. 1968;163(1):229–38.

40. Gachon F, Olela FF, Schaad O, Descombes P, Schibler U. The

circadian PAR-domain basic leucine zipper transcription factors

DBP, TEF, and HLF modulate basal and inducible xenobiotic

detoxification. Cell Metab. 2006;4(1):25–36.

41. Lu J-F, Eppler SM, Wolf J, et al. Clinical pharmacokinetics of

erlotinib in patients with solid tumors and exposure-safety rela-

tionship in patients with non-small cell lung cancer. Clin Phar-

macol Ther. 2006;80(2):136–45.

42. Li J, Karlsson MO, Brahmer J, et al. CYP3A phenotyping

approach to predict systemic exposure to EGFR tyrosine kinase

inhibitors. J Natl Cancer Inst. 2006;98(23):1714–23.

43. Bow EJ, Kilpartick MG, Scott BA, Clinich JJ, Cheang MS. Acute

myeloid leukemia in Manitoba. Cancer. 1994;74(1):52–60.

44. Bow EJ, Loewen R, Cheang MS, Shore TB, Rubinger M, Sch-

acter B. Cytotoxic therapy-induced D-xylose malabsorption and

invasive infection during remission-induction therapy for acute

myeloid leukemia in adults. J Clin Oncol. 1997;15(6):2254–61.

45. Johnson E, MacGowan A, Potter M, et al. Reduced absorption of

oral ciprofloxacin after chemotherapy for haematological malig-

nancy. J Antimicrob Chemother. 1990;25(5):837–42.

46. Sitar DS, Aoki FY, Bow EJ. Acyclovir bioavailability in patients

with acute myelogenous leukemia treated with daunorubicin and

cytarabine. J Clin Pharmacol. 2008;48(8):995–8.

47. Bow E, Meddings J. Intestinal mucosal dysfunction and infection

during remission-induction therapy for acute myeloid leukaemia.

Leukemia. 2006;20(12):2087–92.

48. Klotz U. Pathophysiological and disease-induced changes in drug

distribution volume: pharmacokinetic implications. Clin Phar-

macokinet. 1976;1(3):204–18.

49. Gupta N, Chiu Y, Toh H, et al. Preliminary pharmacokinetics and

safety comparison of Child–Pugh A (CPA) vs Child–Pugh B

(CPB) patients enrolled in a phase 2 study in hepatocellular

carcinoma (HCC). Presented at Annals of Oncology; 2010.

50. Frohna P, Lu J, Eppler S, et al. Evaluation of the absolute oral

bioavailability and bioequivalence of erlotinib, an inhibitor of the

epidermal growth factor receptor tyrosine kinase, in a random-

ized, crossover study in healthy subjects. J Clin Pharmacol.

2006;46(3):282–90.

51. Gandhi M, Aweeka F, Greenblatt RM, Blaschke TF. Sex differ-

ences in pharmacokinetics and pharmacodynamics. Annu Rev

Pharmacol Toxicol. 2004;44:499–523.

PopPK Analysis of Linifanib in Cancer Patients 359


Recommended