Download ASCO 2011 NKTR-102 PopPK Poster

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Pharmacogenomics wikipedia , lookup

Plateau principle wikipedia , lookup

Discovery and development of cyclooxygenase 2 inhibitors wikipedia , lookup

Ofloxacin wikipedia , lookup

Adherence (medicine) wikipedia , lookup

Hormesis wikipedia , lookup

Bad Pharma wikipedia , lookup

Pharmacokinetics wikipedia , lookup

Theralizumab wikipedia , lookup

Dydrogesterone wikipedia , lookup

Bilastine wikipedia , lookup

Transcript
Population Pharmacokinetics of NKTR-102, a Topoisomerase I Inhibitor-Polymer Conjugate in Patients With Advanced Solid Tumors
Michael A. Eldon and Ute Hoch, Nektar Therapeutics, San Francisco, CA
Abstract
NKTR-102 Pharmacokinetic Analysis
Covariate Analysis
Background: NKTR-102 demonstrates significant anti-tumor activity with a tolerable toxicity profile in pts with platinumresistant/refractory ovarian or metastatic breast cancers. NKTR-102 is a novel topoisomerase I inhibitor that uses Nektar’s
proprietary polymer conjugation to improve the PK of its active metabolite SN38 resulting in significantly reduced Cmax and
sustained exposure throughout the dosing interval. Methods: NKTR-102 was administered as a 90-min IV inf. to 94 pts: 76 pts
at 58-245 mg/m2 wx3q4w, q14d or q21d, and 18 pts at 100 or 125 mg/m2 q21d receiving concurrent cetuximab. Serial plasma
samples were collected for the duration of treatment and assayed by LC-MS/MS for NKTR-102, irinotecan, SN38,
SN38-Glucuronide (SN38-G), and APC. Results: NKTR-102 and its metabolites followed biphasic disposition kinetics with
first-order appearance rates consistent with their metabolic progression (Tmax 1.5-24 hr) and first-order terminal t1/2 between
21-61 days. PK of NKTR-102 and all metabolites were independent of dose and schedule. BSA was a significant covariate for
CL. SN38 CL was lower (30%) in females and lower (30%) in pts who were homozygous for UGT1A1*28, but these reductions
were not considered clinically important. Patient age, hepatic or renal functions were not correlated with SN38 CL.
Coadministration of cetuximab had no effects on NKTR-102 PK and vice versa. PK in pts with ovarian and breast cancer were
similar to those with non-ovarian or -breast tumors. The RP2D (145 mg/m2) results in approx. the same plasma SN38 AUC as
350 mg/m2 of irinotecan, but exposure is continuous rather than intermittent and Cmax is 10-fold less. Conclusions: NKTR-102
results in sustained systemic exposure to SN38 between infrequent doses while preventing exposure to excessively high
concentrations; these enhancements appear responsible for improved efficacy and tolerability profiles. The PK of NKTR-102 and
metabolites are predictable and do not require complex dosing adjustments and support ongoing Phase 2 clinical development as
single-agent in metastatic breast, platinum-resistant ovarian, and metastatic colorectal cancers.
Observed and Model-Predicted Concentration-Time Profiles for Representative Patients at the Recommended Phase 2 Doses
Listing of Potential and Statistically Significant Covariates
100000
100000
100000
Plasma Analyte Conc. (ng/mL)
1000000
100
10
100
100
100
1
0.1
10
10
10
1
1
1
0.1
0.1
0
5
10
15
20
25
0
5
10
Time (weeks)
15
20
0.1
25
0
5
Time (weeks)
10
0
Time (weeks)
15
Symbols = observed
concentrations
Lines = model-predicted
concentrations
1
20
Time (weeks)
AUC
(ng*hr/mL)
50-600
3000-12000
27 (13)
SN38
76
13 (170)
2-6
400-2500
50 (28)
SN38-G
76
16 (69)
30-100
6500-67000
61 (37)
APC
76
20 (41)
4-30
800-3000
53 (8)
–
Cmax of NKTR-102 observed shortly after the end of the 90-min infusion
–
Cmax decreased in the order of NKTR-102>irinotecan≥SN38-G>APC>SN38
–
Tmax for NKTR-102 metabolites increased, consistent with the proposed metabolic scheme
–
NKTR-102 and metabolite PK were independent of dose and treatment schedule, and Cmax and AUC
increased linearly with dose
Plasma SN38 Conc. (ng/mL)
Plasma Irinotecan Conc. (ng/mL)
60
80
100
0.0
20
40
60
80
100
1
400
100000
300
90%
100
80%
0
100%
101%
100%
0.15
106%
0.05
0
0.00
103%
3
6
9
Time (weeks)
10
400000.0
200000.0
40
60
80
PK in patients with ovarian cancer is similar to those with all other cancers
0.0
100
–
0
20
40
60
80
100
0.0
–
40
60
n=33
80
100
n=50
2000000.0
20000
1000000.0
100%
80%
< 65
≥ 65
500000.0
0.0
0.15
80
60
100%
100%
100%
0
≥ 65
< 65
126%
≥ 65
Small but statistically significant reduction observed in irinotecan clearance between
the age groups (ages ranged from 25-81 years)
Small decline is not expected to be clinically important in the selection of NKTR-102
starting dose
1.5×1008
600000.0
n=45
1.0×1008
400000.0
200000.0
100%
70%
15
100%
90%
5.0×1007
100%
0
SN38 CL/Fm is ~30% less in females
–
No other parameter significantly influenced by patient gender
600000.0
20
100%
15
70%
400000.0
0.0
AUCdose normalized
1.0×10 08
100%
n=11
5.0×10 07
100%
130%
10
5
70%
0
0.0
–
Reduced SN38 Cmax and overall lower SN38 plasma concentrations appear less
likely to result in saturation of glucuronidation in homozygous patients
Reduction in starting dose not expected to be required
60
PK in patients with breast cancer is similar to those with all other cancers
80
100
–
0.00
76%
0
–
Only SN38-G formation rate (Kform) found to be significantly different
–
Consistent with role of UGT enzymes in SN38 to SN38-G metabolism
–
This finding is not expected to be clinically important for the selection of
NKTR-102 starting dose
Conclusions
• SN38 concentrations are sustained throughout a 21-day dosing
interval; Cmax ~5-10-fold less for NKTR-102 vs. irinotecan
V/Fm
n=83
100%
• NKTR-102 and metabolites demonstrate dose proportional
disposition kinetics with long disposition half-lives
UGT1A1 Status and SN38 PK
1.5×10 08
65%
100
10
–
800000.0
200
120%
5
0.0
0.0
n=11
100%
20
n=49
5
40
≥ 65
AUCdose normalized
V/Fm
Reductions in clearance and volume in homozygous patients were not significant
20
65%
AUCdose normalized
300
0.05
20
–
0
< 65
40
< 65
800000.0
n=83
0.10
10
0
100%
Kform
AUCdose normalized
2500000.0
1500000.0
15
20
≥ 65
0.00
UGT1A1 Status and SN38G PK
V/Fm
30000
20
0
< 65
80%
Clinical significance of these trends, if any, is unknown
SN38 AUCdose normalized
5.0×10 07
100%
–
Clearance
1.0×10 08
n=36
0.05
Renal excretion of NKTR-102 constitutes only a minor pathway; predominant
metabolic pathway is dependent on cleavage to irinotecan
Due to the small changes in clearance, not expected that patients with moderate
renal impairment (eGFR ≥30 mL/min) will require reduced NKTR-102 starting doses
Clearance
1.5×10 08
n=58
–
Gender and SN38 PK
SN38 V/Fm
0.10
Kform, CL, and V/Fm for SN38-G and for APC all appear to decline with age,
but only Kform was statistically significant
–
12
n=36
0.10
50000
0.15
n=58
116%
APC Kform
–
145 mg/m2
NKTR-102
0.1
SN38 Clearance
600000.0
20
100%
200000.0
15
0
150000
500
n=22
SN38-G Kform
AUCdose normalized
Only NKTR-102 was observed to decrease with decreasing renal function
–
Five of 94 patients had breast as the primary tumor. Patients with breast cancer had
similar characteristics to those with other cancers.
800000.0
0
V1
–
350 mg/m2
Irinotecan
Compared to irinotecan administration, SN38 concentrations are sustained between dosing
occasions after NKTR-102 administration: plasma SN38 remains >0.4 ng/mL after the first
dose and > 1 ng/mL after ≥4 doses
– In contrast, plasma SN38 concentrations fall below 0.1 ng/mL 7 days post irinotecan dose,
resulting in a drug holiday for 70% of the dosing cycle
20
5
0
Clearance
Age and SN38-G PK or APC PK
UGT1A1*28
40
not investigated
Female
20
ka, CL, V/Fm
NonUGT1A1*28
–
0
CL, V/Fm
Male
0.0
Kform, CL, V/Fm
UGT1A1*28
200000.0
Kform, CL, V/Fm
Female
5.0×10 07
APC
NonUGT1A1*28
400000.0
Kform, CL, V/Fm
UGT1A1*28
10
–
SN38 AUC dose normalized
1.0×10 08
CL, V/Fm
UGT1A1*28
600000.0
CL, V/Fm
200
10000
– A single145 mg/m2 dose of NKTR-102 results in approximately the same plasma exposure to
SN38 as the 350 mg/m2 dose of irinotecan, but exposure is continuous rather than intermittent
and maximal concentrations are ~5- to 10-fold less
Six of 94 patients had ovarian as the primary tumor. Patients with ovarian cancer tended
to be smaller (mean BSA 1.72 vs 1.91 m2), older (mean age 65 vs 58 yr) and have
more renal impairment (mean eGFR 53 vs 79 mL/min) than those with other cancers.
1.5×10 08
Kform, CL, V/Fm
40000
PK in Patients With Ovarian and Breast Cancers (Phase 3 planning underway for these tumor types)
800000.0
Kform, CL, V/Fm
Male
Reduced Cmax is likely associated with lack of cholinergic reactions after
administration of NKTR-102
SN38 V/Fm
SN38-G
Female
Single agent approved dose of irinotecan is 350 mg/m q21d
– Compared to irinotecan administration, irinotecan Cmax after administration of
145 mg/m2 NKTR-102 is ~35-fold lower compared to administration of 350 mg/m2
irinotecan
2
SN38 Clearance
CL, V/Fm
n=43
50000
Irinotecan and SN38 concentrations after irinotecan administration are based on Xie et al. 2002, JCO 20:3293. Simulations of irinotecan and SN38 after NKTR-102 administration are based on the 06-IN-IR001 derived population parameters.
–
CL, V/Fm
30
0
0
–
CL, V/Fm
Clearance
Predicted SN38 Concentrations
12
Time (weeks)
Kform, CL, V/Fm
NonUGT1A1*28
9
Kform, CL, V/Fm
UGT1A1*28
1
6
SN38
NonUGT1A1*28
Covariate Analysis: The effect of patient demographics, renal or hepatic impairment as estimated from clinical laboratory
results and UGT1A1 homozygosity on drug and metabolite CL or CL/F, V or V/F, and metabolite formation rate (Kform) were
investigated using nonlinear mixed-effects modeling. Plasma concentration-time data for NKTR-102, irinotecan, SN38,
SN38-G, or APC from 94 patients (76 receiving single agent NKTR-102; 18 receiving concurrent cetuximab; data were pooled
because no drug-drug interaction was observed in the presence of cetuximab) with various solid tumors were combined for
analysis. Individual patient concentration-time profiles for all analytes were well represented by a 2-compartment
pharmacokinetic model with either zero-order input for NKTR-102 or first-order input for all metabolites, and first-order output
for all analytes. Data for each analyte were fitted with the appropriate PK model and selected clinical covariates using Monolix
3.1. Inclusion of a covariate in the final model for each analyte was based on application of Wald’s approximation of the
likelihood ratio test as described by Kowalski and Hutmacher (Efficient screening of covariates in population models using
Wald's approximation to the likelihood ratio test. J Pharmacokinet Pharmacodyn 2001 Jun;28(3):253-75).
145 mg/m2
NKTR-102
3
not investigated
Male
NKTR-102 Pharmacokinetic Analysis: In Phase 1 Study 06-IN-IR001, NKTR-102 was administered as a 90-min IV infusion.
Thirty-two patients received 58-230 mg/m2 doses wx3q4wk, 19 pts received 145-230 mg/m2 q14d, and 25 pts received 145-245
mg/m2 doses q21d. Plasma samples were obtained throughout the treatment duration: rich PK sampling was performed during
the first cycle and cycle 3 for the q14d and q21d treatment schedules; predose samples were obtained before each subsequent
cycle. PK samples were analyzed using validated methods with LLOQ of 2000 ng/mL for NKTR-102, 1 ng/mL for irinotecan,
SN38-G, and APC, and 0.2 ng/mL for SN38. Pharmacokinetics of all analytes were estimated using nonlinear mixed-effects
modeling. Individual patient concentration-time profiles for all analytes were well represented by a 2-compartment
pharmacokinetic model with either zero-order input for NKTR-102 or first-order input for all metabolites, and first-order output for
all analytes. Data for each analyte were fitted with the appropriate PK model using Monolix 3.1 (INRIA Saclay, Orsay, France).
350 mg/m2
Irinotecan
0
CL, V/Fm
Age and Irinotecan PK
10000
0.1
n=29
40
0
Interpatient variability in SN38 exposure after NKTR-102 was similar to that reported for SN38 after
irinotecan dministration
–
Predicted Irinotecan Concentrations
10
CL, V/Fm
Non UGT1A1*28
SN38-Glucuronide
(inactive)
CL, V/Fm
10
*Data from 9 pts were missing due to incorrect sample processing.
100
CL, V/Fm
Moderate
4 (91)
1000
Irinotecan
Mild
67*
UGT1A1
not investigated
Normal
21 (20)
Irinotecan
SN38
CL, V1
Moderate
1.8 (31)
CL, V1
20
T1/2
(days)
5600000 29000000
CL, V1
Mild
3600001300000
CL, V1
Normal
76
Cmax
(ng/mL)
NKTR-102*
Renal Impairment and NKTR-102 PK
Mean (%CV) plasma pharmacokinetic parameters for NKTR-102, irinotecan, SN38, SN38-G, and APC after a 90-min intravenous infusion of the first NKTR-102 dose between 58-245 mg/m
Tmax
(hr)
UGT1A1*28
11 of 94 pts
25
2
n
Bilirubin
0.2-1.6 mg/dL
#Estimated glomerular filtration rate based on corrected chronic kidney disease epidemiological models
*Black color indicates covariates investigated, red color indicates the covariates found statistically significant
PK Parameters
Methods
Abstract 2598, Poster 8E ASCO 2011, Developmental Therapeutics – Clinical Pharmacology and Immunotherapy, June 3-7, 2011
To download a copy of this poster, please visit http://www.nektar.com/
1000
Reduced UGT1A1 Activity
eGFR#
30-134 mL/min
Moderate
APC, NPC
(inactive)
1000
Hepatic Impairment
Gender
45 female, 49 male
Mild
CYP3A4
10000
Renal Impairment
Age
25-81 years
Normal
Irinotecan
10000
1000
1000
NKTR-102
Irinotecan
SN38
SN38-G
APC
1000000
100000
10000
10000
Demographics
Comparison of Irinotecan and SN38 Concentration-Time Profiles after NKTR-102 and Irinotecan Administration
Esterases
NKTR-102
Pt 1020, 145 mg/m2 q21d
1000000
NKTR-102
-- NKTR-102 uses Nektar’s proprietary polymer conjugation to improve the pharmacokinetics of irinotecan and its active
metabolite SN38.
-- NKTR-102 has superior activity compared to irinotecan in a wide range of mouse models of human xenograft tumors.
-- NKTR-102 demonstrated high anti-tumor activity in a range of tumors in Phase 1 (11% confirmed PRs; ENA 2008, abs 595).
-- NKTR-102 showed a 22% confirmed response rate per RECIST in heavily pre-treated women with platinum resistant/refractory
ovarian cancer (ASCO 2010, abs 5013), and a 29% confirmed response rate in 2nd/3rd line patients with advanced breast
cancer (SABCS 2010, abs S6-6)
Pt 1022, 145 mg/m2 q14d
1000000
Analyte
Background
Irinotecan is extensively metabolized to various metabolites.
It is cleaved enzymatically to form SN38 which is conjugated
to an inactive glucuronide, SN38-G. Other inactive irinotecan
metabolites identified are the major plasma metabolite APC
(7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin), and NPC (7-ethyl-10-[4-amino-1piperidino]-carbonyloxycamptothecin). In NKTR-102, irinotecan
is conjugated to a multi-arm PEG and is cleaved in vivo.
Pt 2004, 115 mg/m2 wx3q4wk
• Interpatient variability in PK parameters is comparable to
irinotecan
• No unexpected clinical covariates of drug/metabolite exposure
• NKTR-102 appears to allow for a uniform starting dose across
all patients
• Overall, the PK of NKTR-102 and its metabolites are predictable
and do not require complex dosing adjustments
• NKTR-102 is currently being developed in ovarian, breast, and
colorectal cancers and Phase 3 planning is underway for
patients with ovarian and breast cancers