+ All Categories
Home > Documents > Dogs Are More Sensitive to Antagonists of Inhibitor of Apoptosis Proteins Than Rats and Humans: A...

Dogs Are More Sensitive to Antagonists of Inhibitor of Apoptosis Proteins Than Rats and Humans: A...

Date post: 01-May-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
9
Dogs Are More Sensitive to Antagonists of Inhibitor of Apoptosis Proteins Than Rats and Humans: A Translational Toxicokinetic/Toxicodynamic Analysis Harvey Wong,* ,1 Nageshwar R. Budha,Kristina West,Elizabeth Blackwood,Joseph A. Ware,Ron Yu,§ Walter C. Darbonne,Stephen E. Gould,Ronald Steigerwalt,|| Rebecca Erickson,|| Cornelis E. A. C. Hop,* Patricia LoRusso,||| S. Gail Eckhardt,|||| Andrew Wagner,# Iris T. Chan,** Michael Mamounas,†† John A. Flygare, a and Wayne J. Fairbrother b *Department of Drug Metabolism and Pharmacokinetics, Department of Clinical Pharmacology, Department of Translational Oncology, §Department of Biostatistics, Department of PD Biomarkers, and ||Department of Safety Assessment, Genentech, Inc., South San Francisco, California; |||Karmanos Cancer Institute, Detroit, Michigan; ||||University of Colorado Cancer Center, Denver, Colorado; #Dana-Farber Cancer Institute, Boston, Massachusetts; and **Oncology Exploratory Clinical Development, ††Portfolio Management and Operations, a Medicinal Chemistry, and b Early Stage Discovery Biochemistry, Genentech, Inc., South San Francisco, California 1 To whom correspondence should be addressed at Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, MS 412a, South San Francisco, CA 94080. Fax: (650) 467-3487. E-mail: [email protected]. Received June 11, 2012; accepted July 24, 2012 Inhibitor of apoptosis (IAP) proteins suppress apoptosis and are overexpressed in a variety of cancers. GDC-0152 is a potent and selective IAP antagonist being developed as an anticancer agent. In preclinical safety studies, dogs were particularly sensitive to GDC- 0152 showing adverse signs of a tumor necrosis factor alpha (TNF- α) driven systemic inammatory response, related to cellular IAP degradation and activation of NFκB signaling, at lower exposures compared with rat. In addition, downstream increases in systemic levels of cytokines and chemokines, such as monocyte chemotactic protein-1 (MCP-1), were observed. A semimechanistic population toxicokinetic/toxicodynamic (TK/TD) model incorporating transit compartments was used to t MCP-1 plasma concentrations from rats or dogs given iv GDC-0152 doses. Estimated TD parameters inferred that lower GDC-0152 plasma concentrations triggered more severe increases in plasma MCP-1 in dogs compared with rats. Human simulations performed using dog TD parameters and human pharmacokinetics predicted 300–2400% increases of MCP-1 in humans at iv doses from 0.76 to 1.48 mg/kg. Similar simulations using rat TD parameters suggest little or no change. Patients given iv doses of GDC-0152 up to 1.48 mg/kg iv showed no substantial increases in systemic MCP-1 or signs of a severe TNF-α driven systemic inammatory response. Emerging clinical data reported for other IAP antagonists are consistent with our observations. Taken together, the data suggest dogs are more sensitive to IAP antagonists compared with humans and rats. This study illustrates how TK/TD analysis can be utilized to quantitatively translate and context an identied preclinical safety risk in dogs to humans. Key Words: TK/TD; PK/PD; pharmacokinetic-pharmacody- namic; toxicodynamics; toxicokinetics; IAP. Inhibitor of apoptosis (IAP) proteins are involved in the regulation of programmed cell death or apoptosis. They were rst identied in baculoviruses where they function to sup- press host cell death responses during viral infection (Flygare and Fairbrother, 2010; Varfolomeev and Vucic, 2011). IAP proteins suppress apoptosis, in part, by inhibiting activated cytosolic cysteine/aspartate-specic proteases (caspases) that are critical for its execution (Flygare and Fairbrother, 2010; Varfolomeev and Vucic, 2011). The mammalian family of IAP proteins includes X-linked IAP (XIAP), cellular IAP 1 and 2 (cIAP1 and cIAP2), and melanoma IAP (ML-IAP). Of the IAP proteins, only XIAP directly inhibits effector caspases-3 and -7, which execute the apoptotic death pro- gram. XIAP also indirectly inhibits effector-caspase activ- ity via inhibition of the initiator caspase-9. ML-IAP and cIAPs directly antagonize second mitochondria-derived activator of caspase/direct IAP-binding protein with low pI (Smac/DIABLO), an endogenous IAP antagonist that acts by blocking XIAP-mediated inhibition of caspases. cIAPs have an additional role of preventing death receptor (e.g., tumor necrosis factor alpha [TNF-α], Fas, DR4, and DR5) mediated activation of initiator caspase-8, thereby stop- ping downstream activation of effector caspases-3 and -7. Finally, cIAPs positively regulate canonical NF-κB signal- ing and negatively regulate noncanonical NF-κB signaling and induction of NF-κB target genes, including TNF-α. IAP proteins are frequently overexpressed in cancer cells where they serve as regulators of cancer cell survival and are often indicators of poor prognosis (Vucic and Fairbrother, 2007). Conversely, inhibition of IAP proteins has been shown to sen- sitize cancer cells to proapoptotic anticancer agents. Thus, Disclaimer: The authors certify that all research involving human subjects was done under full compliance with all government policies and the Helsinki Declaration. toxicological sciences 130(1), 205–213 (2012) doi:10.1093/toxsci/kfs235 Advance Access publication July 27, 2012 © The Author 2012. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For permissions, please email: [email protected] by guest on October 17, 2012 http://toxsci.oxfordjournals.org/ Downloaded from
Transcript

Dogs Are More Sensitive to Antagonists of Inhibitor of Apoptosis Proteins Than Rats and Humans: A Translational Toxicokinetic/Toxicodynamic

Analysis

Harvey Wong,*,1 Nageshwar R. Budha,† Kristina West,‡ Elizabeth Blackwood,‡ Joseph A. Ware,† Ron Yu,§ Walter C. Darbonne,¶ Stephen E. Gould,‡ Ronald Steigerwalt,|| Rebecca Erickson,|| Cornelis E. A. C. Hop,* Patricia LoRusso,|||

S. Gail Eckhardt,|||| Andrew Wagner,# Iris T. Chan,** Michael Mamounas,†† John A. Flygare,a and Wayne J. Fairbrotherb

*Department of Drug Metabolism and Pharmacokinetics, †Department of Clinical Pharmacology, ‡Department of Translational Oncology, §Department of Biostatistics, ¶Department of PD Biomarkers, and ||Department of Safety Assessment, Genentech, Inc., South San Francisco, California; |||Karmanos

Cancer Institute, Detroit, Michigan; ||||University of Colorado Cancer Center, Denver, Colorado; #Dana-Farber Cancer Institute, Boston, Massachusetts; and **Oncology Exploratory Clinical Development, ††Portfolio Management and Operations, aMedicinal Chemistry, and bEarly Stage Discovery Biochemistry,

Genentech, Inc., South San Francisco, California

1To whom correspondence should be addressed at Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, MS 412a, South San Francisco, CA 94080. Fax: (650) 467-3487. E-mail: [email protected].

Received June 11, 2012; accepted July 24, 2012

Inhibitor of apoptosis (IAP) proteins suppress apoptosis and are overexpressed in a variety of cancers. GDC-0152 is a potent and selective IAP antagonist being developed as an anticancer agent. In preclinical safety studies, dogs were particularly sensitive to GDC-0152 showing adverse signs of a tumor necrosis factor alpha (TNF-α) driven systemic inflammatory response, related to cellular IAP degradation and activation of NFκB signaling, at lower exposures compared with rat. In addition, downstream increases in systemic levels of cytokines and chemokines, such as monocyte chemotactic protein-1 (MCP-1), were observed. A  semimechanistic population toxicokinetic/toxicodynamic (TK/TD) model incorporating transit compartments was used to fit MCP-1 plasma concentrations from rats or dogs given iv GDC-0152 doses. Estimated TD parameters inferred that lower GDC-0152 plasma concentrations triggered more severe increases in plasma MCP-1 in dogs compared with rats. Human simulations performed using dog TD parameters and human pharmacokinetics predicted 300–2400% increases of MCP-1 in humans at iv doses from 0.76 to 1.48 mg/kg. Similar simulations using rat TD parameters suggest little or no change. Patients given iv doses of GDC-0152 up to 1.48 mg/kg iv showed no substantial increases in systemic MCP-1 or signs of a severe TNF-α driven systemic inflammatory response. Emerging clinical data reported for other IAP antagonists are consistent with our observations. Taken together, the data suggest dogs are more sensitive to IAP antagonists compared with humans and rats. This study illustrates how TK/TD analysis can be utilized to quantitatively translate and context an identified preclinical safety risk in dogs to humans.

Key Words: TK/TD; PK/PD; pharmacokinetic-pharmacody-namic; toxicodynamics; toxicokinetics; IAP.

Inhibitor of apoptosis (IAP) proteins are involved in the regulation of programmed cell death or apoptosis. They were first identified in baculoviruses where they function to sup-press host cell death responses during viral infection (Flygare and Fairbrother, 2010; Varfolomeev and Vucic, 2011). IAP proteins suppress apoptosis, in part, by inhibiting activated cytosolic cysteine/aspartate-specific proteases (caspases) that are critical for its execution (Flygare and Fairbrother, 2010; Varfolomeev and Vucic, 2011). The mammalian family of IAP proteins includes X-linked IAP (XIAP), cellular IAP 1 and 2 (cIAP1 and cIAP2), and melanoma IAP (ML-IAP). Of the IAP proteins, only XIAP directly inhibits effector caspases-3 and -7, which execute the apoptotic death pro-gram. XIAP also indirectly inhibits effector-caspase activ-ity via inhibition of the initiator caspase-9. ML-IAP and cIAPs directly antagonize second mitochondria-derived activator of caspase/direct IAP-binding protein with low pI (Smac/DIABLO), an endogenous IAP antagonist that acts by blocking XIAP-mediated inhibition of caspases. cIAPs have an additional role of preventing death receptor (e.g., tumor necrosis factor alpha [TNF-α], Fas, DR4, and DR5) mediated activation of initiator caspase-8, thereby stop-ping downstream activation of effector caspases-3 and -7. Finally, cIAPs positively regulate canonical NF-κB signal-ing and negatively regulate noncanonical NF-κB signaling and induction of NF-κB target genes, including TNF-α. IAP proteins are frequently overexpressed in cancer cells where they serve as regulators of cancer cell survival and are often indicators of poor prognosis (Vucic and Fairbrother, 2007). Conversely, inhibition of IAP proteins has been shown to sen-sitize cancer cells to proapoptotic anticancer agents. Thus,

Disclaimer: The authors certify that all research involving human subjects was done under full compliance with all government policies and the Helsinki Declaration.

toxicological sciences 130(1), 205–213 (2012)doi:10.1093/toxsci/kfs235Advance Access publication July 27, 2012

© The Author 2012. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved. For permissions, please email: [email protected]

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

the development of IAP antagonists as potential anticancer agents has been of interest.

GDC-0152 (Fig.  1) is a potent pan inhibitor of IAP pro-teins with binding affinities to the BIR3 domains of X-IAP and cIAP1/2, and the BIR domain of ML-IAP in the low nano- molar range (Ki < 50nM) using a fluorescence polarization-based competition assay (Flygare et  al., 2012). GDC-0152 caused a decrease in cell viability in MDA-MB-231 human breast cancer cells while having no effect on normal human mammary epithe-lial cells. In MDA-MB-231 xenograft mice studies, GDC-0152 showed robust single-agent activity resulting in significant tumor growth inhibition at weekly oral doses of 10, 50, or 100 mg/kg. GDC-0152 was evaluated in rat safety studies following iv doses of 3–45 mg/kg given once every 2 weeks for 6 weeks resulting in daily exposures (i.e., area under the plasma concentration-time curve [AUC0–∞]) ranging from 3.51 to 113µM·h. Dog safety stud-ies were performed at lower iv doses ranging from 0.3 to 10 mg/kg which accordingly resulted in lower GDC-0152 exposures compared with rat ranging from 0.723 to 16.5µM·h (Erickson et al., 2011). In both rat and dog safety studies, in-life obser-vations included elevated plasma cytokines and chemokines (including TNF-α and monocyte chemotactic protein-1 [MCP-1]), an inflammatory leukogram, and increased liver transami-nases, in addition to histopathological findings of inflammatory infiltrates and apoptosis and/or necrosis in multiple tissues (Erickson et al., 2011). In particular, dogs appeared more sen-sitive to TNF-α driven inflammatory response-associated effects of IAP inhibition with some dogs showing transient clinical signs of fever, tremors, and hypotension that were not as observed in rats (Erickson, personal communication). From these stud-ies, MCP-1 was identified to be the most sensitive downstream marker of TNF-α upregulation and NF-κB activation.

The objective of this study was to characterize species differences in sensitivity to inhibition of IAP proteins using plasma MCP-1, an NF-κB target gene that is downstream of TNF-α, as a marker of a systemic TNF-α driven inflammatory response. More importantly, we utilize toxicokinetic/toxicodynamic (TK/TD) modeling and human data from a GDC-0152 clinical trial to understand how preclinical

safety findings translate to humans. We found that dogs were extremely sensitive to IAP inhibition compared with humans and rats. This study shows an approach utilizing TK/TD modeling to quantitatively understand and properly context preclinical toxicity findings and will aid in the interpretation of human safety risks of IAP antagonists in general.

MATERIALS AND METHODS

Rat studies. Female Sprague Dawley rats (weighting 230–250 g; Charles River, Wilmington, MA) were given an iv dose of 20 (n  =  4), 40 (n  =  4), 80 (n = 4), or 120 (n = 4) mg/kg of GDC-0152 (Genentech Inc., South San Francisco, CA) in 10% hydroxypropyl-β-cyclodextrin and 20mM sodium suc-cinate (pH 5.5) via a femoral vein cannulae. Blood samples (~0.2 ml per sam-ple) were collected from each animal via jugular vein cannulae at the following timepoints: predose, 0.083, 0.25, 1, 3, 6, 9, 12, 24, 36, and 48 h postdose. All samples were collected into tubes containing potassium EDTA as an anticoagu-lant. Blood samples were centrifuged within 30 min of collection and plasma was harvested. Plasma samples were stored at ~−70°C until analysis for GDC-0152 concentrations by a liquid chromatographic-tandem mass spectrometric (LC/MS/MS) assay method. Plasma MCP-1 concentrations were determined by multiplexed sandwich immunoassay using Luminex Rodent Multi-Analyte Profiling (MAP) platform (Rules-Based Medicine, San Antonio, TX).

Dog studies. Male and female beagle dogs (weighing 7–12 kg; Covance Research Products, Cumberland, VA) were given 30-min iv infusions of 0.3 (n = 8), 0.75 (n = 8), 1.5 (n = 6), 5 (n = 6), 10 (n = 6), and 15 mg/kg (n = 6) of GDC-0152 in 15% hydroxypropyl-β-cyclodextrin and 20mM succinic acid in 5% dextrose. Blood samples (~0.8 ml per sample) were collected from each animal via jugular vein cannulae at the following timepoints: predose, 0.083, 0.17, 0.5, 1, 1.5, 3, 6, 12, and 24 h postdose. All samples were collected into tubes containing potassium EDTA as an anticoagulant and kept on ice. Blood samples were centrifuged within 1 h of collection and plasma was harvested. Plasma samples were stored at ~−70°C until analysis for GDC-0152 con-centrations by LC/MS/MS. MCP-1 levels in plasma were measured using an immunoassay (Duoset Canine CCL2 [JE; MCP-1]) ELISA Development Kit (R&D Systems, Minneapolis, MN).

Human clinical study. The human pharmacokinetic (PK) parameters were determined using data from a phase 1a study in cancer patients as described by Flygare et al. (2012). The protocol for the phase 1a study was approved by the Institutional Review Boards at participating institutions. Written informed con-sent was obtained from all trial participants. Briefly, doses ranging from 0.049 to 1.48 mg/kg were administered by a 0.5-h iv infusion once every 14  days to cancer patients. Plasma samples were collected at predose, 0.2, 0.4, 0.8, 1.5, 3, 4.5, 6.5, 9.5, 12.5, 18.5, and 24.5 h postdose. Plasma samples were stored at ~−70°C until analysis for GDC-0152 concentrations by LC/MS/MS. Plasma MCP-1 concentrations were determined using a multiplexed sandwich immunoassay using Luminex Human MAP platform (Rules Based Medicine).

TK analysis. Standard TK parameters (presented in Tables 1 and 2) were calculated by noncompartmental methods as described by Gilbaldi and Perrier (1982). TK parameters are reported as the mean ± SD.

TK/TD modeling. A two-compartment model as described by Equations 1 and 2 (Fig. 2A) was used to characterize the toxicokinetics of GDC-0152 in dogs.

dXdt

k X k X k Xcpc p cp c e c= − −

(1)

dX

dtk X k Xpcp c pc p= −

(2)

where Xc is the amount of GDC-0152 in the central compartment, Xp the amount of GDC-0152 in the peripheral compartment, t the time, kcp the rate constant

FIG. 1. Chemical structure of GDC-0152.

206 WONG ET AL.

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

from central compartment to peripheral compartment, kpc the rate constant from peripheral compartment to central compartment, and ke the elimination rate constant.

Because GDC-0152 toxicokinetics in rats is nonlinear, a two-compartment model described by Equations 2 and 3 was used to characterize the toxicokinet-ics of GDC-0152.

dXdt

k X k XV

V K XXc

pc p cp cmax

c m cc= − −

+ (3)

where Vmax is the maximum rate of GDC-0152 elimination and Km (Michaelis-Menten constant) the concentration associated with ½Vmax.

A semimechanistic population TK/TD model incorporating a series of tran-sit compartments was used to characterize the MCP-1 response to GDC-0152 and is described by Equations 4–8 (Fig. 2B).

dTdt

k X T11= −( )c

(4)

dTdt

k T T21 2= −( ) (5)

dTdt

k T T32 3= −( ) (6)

d

dtk E

TV

kc

nMCP-1

MCP-1in out

( )= +

⎝⎜

⎠⎟

⎝⎜⎜

⎠⎟⎟

− ( )1 3 (7)

kkRoutin=0

(8)

where T1, T2, and T3 represent transit compartments delaying the effect of GDC-0152 on MCP-1, MCP-1 the MCP-1 plasma concentration (pg/ml), k the transit rate constant, E a constant relating GDC-0152 concentrations to its effect on the formation rate of MCP-1, Vc the volume of distribution of the central com-partment, T3/Vc the GDC-0152 concentration following the delay by the transit compartments, n the exponent of the power function “E(T3/Vc)

n” that represents the effect of GDC-0152 on the formation rate of MCP-1, kin the rate of forma-tion of MCP-1, kout the rate constant describing the decrease of MCP-1, and Ro the initial or baseline value of MCP-1.

Population TK/TD modeling was performed using S-ADAPT II, an aug-mented version of ADAPT II with population analysis capabilities (Bauer and Guzy, 2004; D’Argenio and Schumitzky, 1997). The described TK/TD model was fit to GDC-0152 and MCP-1 plasma concentrations simultan-eously for all individual dogs or rats at all dose levels. Intersubject variability was assumed to be log-normally distributed and fitted using an exponential variance model. Residual variability error was modeled using an additive and proportional error model for PK and a proportional error model for MCP-1. Population parameter estimates are presented as the estimate followed by the %SE within parentheses.

Pharmacokinetics of GDC-0152 in humans. Human PK parameters were estimated by fitting a three-compartment model, as described by Equations 9–11, to mean human plasma concentration-time data from cancer patients.

dXdt

k X k X k k X k Xcpc p pc p cp cp c e c= + − +( ) −2 2 2 (9)

dX

dtk X k Xpcp c pc p= − (10)

dX

dtk X k Xpcp c pc p

22 2 2= − (11)

where Xc, Xp, t, kcp, kpc, and ke are as defined previously. Xp2 is the amount of GDC-0152 in a second peripheral compartment, kcp2 the rate constant from cen-tral to the second peripheral compartment, and kpc2 the rate constant from the second peripheral compartment to the central compartment.

RESULTS

Toxicokinetics of GDC-0152 in Rats and Dogs

In order to assess the exposure in the rat and dog studies, GDC-0152 toxicokinetics for rats and dogs were assessed and are summarized in Tables 1 and 2, respectively. Plasma clear-ance (CL) was nonlinear in rats ranging from high (approxi-mately rat hepatic blood flow) at a 20-mg/kg dose to moderate (~36% of rat hepatic blood flow) at a 120-mg/kg dose (Table 1). Accordingly, increases in AUC0–∞ in rat trended toward being greater than dose proportional, and the half life (t1/2) progres-sively increased with increasing dose. The volume of distribu-tion at steady-state (Vss) in rats was moderate to high at ~5 l/kg at all doses. In contrast to the rat, the toxicokinetics of GDC-0152 in dogs was reasonably linear, and AUC0–∞ estimates were dose proportional over the dose range tested (0.3–15 mg/kg). CL was moderate in dogs being ~30–50% of hepatic blood flow (Davies and Morris, 1993). Vss in dogs was low to moderate ran-ging from 0.66 to 1.50 l/kg over the dose range tested. Overall, the AUC0–∞ range tested in rats (12.6–206µM·h) was higher than the AUC0–∞ range tested in dogs (0.85–34.3µM·h). Finally, the AUC0–∞ range observed in rats and dogs generally covered the AUC0–∞ range evaluated in previous preclinical safety studies.

TK/TD Modeling

In order to understand the relationship between GDC-0152 plasma concentrations and MCP-1 response in rats and dogs, a population TK/TD model shown in Figure 2 was simultan-eously fit to GDC-0152 and MCP-1 plasma concentration data from rats or dogs. The described nonlinearity in rat toxicokinet-ics necessitated the use of a TK model (Fig. 2A) with nonlinear elimination of GDC-0152. Figures 3A and D show representa-tive concentration-time plots for GDC-0152 and MCP-1 in rat and dog, respectively. A system of transit compartments were used to characterize the delayed MCP-1 response to GDC-0152 observed in both species. Finally, despite higher GDC-0152 concentrations in the rat, the MCP-1 response was substantially higher in the dog (Figures 3A and D ).

Plots of observed versus individual predicted GDC-0152 and MCP-1 plasma concentrations are shown for the rat (Figs. 3B and C) and dog (Figs. 3E and F). As mentioned, representative concentration-time plots of observed GDC-0152 and MCP-1

TABLE 1Toxicokinetics of GDC-0152 in Rats

Dose (mg/kg)

TK parameters

CL (ml/min/kg)

Cmax (µM)

AUC0–∞ (µM·h)

t1/2 (h)

Vss (l/kg)

20 (n = 4) 53.4 ± 5.3 9.16 ± 1.52 12.6 ± 1.3 1.47 ± 0.52 5.57 ± 1.1340 (n = 4) 34.3 ± 4.6 20.2 ± 3.3 39.9 ± 7.0 3.13 ± 1.14 5.29 ± 1.1280 (n = 4) 24.5 ± 3.7 55.3 ± 12.6 111 ± 18 3.99 ± 0.54 5.00 ± 0.76120 (n = 4) 19.9 ± 4.0 92.9 ± 6.0 206 ± 37 4.32 ± 0.88 5.14 ± 0.45

TK/TD ANALYSIS OF MCP-1 207

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

plasma concentrations and model predictions for a representa-tive rat and dog are presented in Figures 3A and D, respectively. Overall, the described integrated TK/TD model adequately described the observed GDC-0152 and MCP-1 plasma concen-tration data for both preclinical species.

TK and TD parameter estimates from rats and dogs following fitting of the integrated TK/TD model are presented in Tables 3 and 4. TK and TD parameters and inter- and intrasubject variances were estimated with good precision for both rats and dogs with standard errors being ≤ 45%. The parameters E and n relate GDC-0152 plasma concentrations to MCP-1 response. The effect of GDC-0152 plasma concentrations on increasing the formation rate of MCP-1 is described in Equation 7 where this effect is equal to “E(T3/Vc)

n” where “T3/Vc” represents GDC-0152 plasma concentrations following the delay created by the transit compartments. The estimate of the typical value of E is ~30-fold higher in dogs compared with rats (Table 4). In addition, the estimate of “n” is greater than twofold higher in the dog. Figure 4 shows a plot describing the relationship between

GDC-0152 plasma concentrations and MCP-1 response. The plot clearly illustrates the MCP-1 response in dogs is more sensitive to IAP inhibition by GDC-0152.

Human Translational MCP-1 TK/TD Modeling

In order to investigate MCP-1 sensitivity to GDC-0152 in humans, the rat and dog TK/TD models and human pharmaco-kinetics were used to simulate predicted MCP-1 responses in humans. PK parameters for humans were estimated by fitting a three-compartment model to individual plasma concentra-tion-time information from a phase 1a study in cancer patients (Flygare et al., 2012). PK parameters are presented in Table 5. A plot of observed versus individual predicted GDC-0152 con-centrations for this fit is presented in Supplementary figure S1.

TD parameters were fixed to values estimated for rat or dog, and simulations were performed using the GDC-0152 PK model for humans to drive the MCP-1 response. Figure 5 shows anticipated percent changes in MCP-1 response in humans for the 0.76, 1.06, and 1.48 mg/kg iv doses based upon rat (Fig. 5A) or dog (Fig. 5B) TD parameters. Based upon rat TD parameters, no dramatic change in MCP-1 was anticipated in humans with a maximum predicted change of < 50% over the dose range evaluated. In contrast, simulations using dog TD parameters suggested a maximum dose-dependent change ran-ging from ~300 to 2400%.

In cancer patients, no dramatic changes in MCP-1 response or signs of a systemic inflammatory response were observed following GDC-0152 administration. Measured maximum percent changes of MCP-1 from the human clinical trial are presented in Figure 6. Based upon the variability observed in MCP-1, there was no significant dose-dependent increase in MCP-1 response to GDC-0152 in humans. These data suggest that human MCP-1 response to IAP inhibition is not as sensi-tive as observed for dogs and is more in line with observations in the rat.

DISCUSSION

Efforts to discover selective inhibitors of IAP proteins in recent years have been accompanied by elucidation of the mech-anisms of their antitumor activity, both as single agents and

FIG. 2. Diagram of population TK/TD structural model. A  two- compartment model was used to describe the toxicokinetics of GDC-0152 (A) A  semimechanistic population TK/TD model incorporating a series of tran-sit compartments was used to characterize the effect of GDC-0152 on MCP-1 levels (B).

TABLE 2Toxicokinetics of GDC-0152 in Dogs

Dose (mg/kg)

TK parameters

CL (ml/min/kg) Cmax (µM) AUC0–∞ (µM·h) t1/2 (h) Vss (l/kg)

0.3 (n = 8) 12.8 ± 3.5 0.630 ± 0.188 0.850 ± 0.276 1.26 ± 0.30 0.665 ± 0.1220.75 (n = 8) 16.0 ± 4.6 1.28 ± 0.35 1.70 ± 0.56 1.67 ± 0.84 0.741 ± 0.1791.5 (n = 6) 15.4 ± 6.4 5.87 ± 6.99 4.01 ± 2.37 0.711 ± 0.026 0.629 ± 0.2865 (n = 6) 11.5 ± 1.8 9.87 ± 2.46 14.9 ± 2.1 1.38 ± 0.41 0.844 ± 0.18010 (n = 6) 15.3 ± 4.3 10.8 ± 3.0 23.3 ± 5.9 1.56 ± 0.40 1.50 ± 0.6215 (n = 6) 15.1 ± 3.0 21.2 ± 7.3 34.3 ± 6.4 1.83 ± 0.05 1.26 ± 0.23

208 WONG ET AL.

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

in combination with other anticancer agents. IAP antagonists induce cIAP protein E3 ubiquitin ligase activity that results in ubiquitination of RIP1, as well as rapid autoubiquitination and proteasomal degradation of the cIAP proteins (Dueber et al., 2011; Varfolomeev et al., 2007; Vince et al., 2007). RIP1 ubiq-uitination induces activation of the canonical NF-κB pathway. The cIAP proteins also regulate the noncanonical NF-κB path-way through ubiquitination of NIK—their loss therefore results

in activation of the noncanonical pathway via NIK stabiliza-tion. NF-κB pathway activation leads to induction of NF-κB target genes which include TNF-α and subsequent downstream cytokine and chemokines. In the absence of cIAP proteins, TNF-α binds to TNFR1 resulting in caspase-8 activation and initiation of apoptosis (Varfolomeev et al., 2007; Vince et al., 2007). Thus, the single-agent activity of IAP antagonists is dependent on TNF-α. More recent data suggest that inhibition

FIG. 3. Representative GDC-0152 or MCP-1 plasma concentration-time plots for a rat given a 120-mg/kg iv dose (A) and dog given a 10-mg/kg iv dose (D). Observed vs. individual predicted GDC-0152 plasma concentrations in rat (B) and dog (E). Observed vs. individual predicted MCP-1 plasma concentrations in rat (C) and dog (F).

TABLE 3TK Parameter Estimates From MCP-1 Population TK/PD Model for Rat and Dog

TK parameters

Rat (n = 16) Dog (n = 40)

Population mean (%SE) Interindividual variance (%SE) Population mean (%SE) Interindividual variance (%SE)

kpc (h−1) 0.195 (44.2) 2.89 (0.706) 4.64 (9.52) 0.105 (37.6)

kcp (h−1) 0.365 (9.30) 0.0879 (13.1) 39.7 (16.5) 0.391 (37.9)

Vc (l/kg) 3.73 (6.22) 0.0460 (19.3) 0.0987 (13.0) 0.257 (30.9)ke (h

−1) — — 9.41 (10.4) 0.170 (40.9)Vmax (µmol/h/kg) 23.5 (14.5) 0.278 (9.13) — —Km (µM) 5.57 (23.5) 0.724 (7.41) — —Residual variability Proportional error (σprop) 0.201 (7.92) — 0.242 (5.64) — Additive error (σadd) 0.00533 (22.7) — 0.00222 (14.2) —

TK/TD ANALYSIS OF MCP-1 209

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

of both cIAP and XIAP proteins is necessary for more efficient single-agent activity of IAP antagonists (Ndubaku et al., 2009). In contrast to single-agent activity, combination activity of IAP antagonists with proapoptotic receptor antagonists appears to be related to the inhibition of XIAP and can be independent of TNF-α signaling (Varfolomeev et al., 2009).

Prior to testing new molecular entities in humans, a thorough preclinical safety evaluation is conducted in a rodent and a non-rodent species. A safe starting dose for first in human studies

is determined based on this evaluation of preclinical safety (DeGeorge et al., 1998; ICH, 2010). The first in human starting dose is usually determined using the most sensitive preclinical species with the safety of human subjects participating in the clinical trial being of the utmost importance. Based upon this paradigm, toxicological evaluations in preclinical species that are particularly sensitive to a new molecular entity’s pharma-cology or off-target toxicity can result in an unnecessarily low starting dose. In the phase 1 oncology setting, this is less than ideal because more patients will be exposed to subtherapeutic doses, and cancer patients primarily participate in such clinical trials with the hope of clinical benefit. Therefore, the appropri-ate translation of preclinical safety risks to humans requires not only an understanding of the relevance of the preclinical safety findings, but also an understanding of the sensitivity of the pre-clinical species to the particular toxicity identified in relation to man.

Our previous work characterizing the toxicology profile of the pan IAP antagonist, GDC-0152, described the link between TNF-α pharmacology and adverse findings in rats and dogs (Erickson et  al., 2011). Dogs exhibited transient increases of TNF-α, MCP-1, IL-6, and IFNγ at relatively low doses compared with rat. These alterations in cytokine and chemokine profiles were consistent with clinical signs (e.g., fever) in dogs and dose-dependent acute systemic inflammatory responses in both rats and dogs. Furthermore, liver injury characterized by dose-dependent increases in serum liver enzymes and/or total bilirubin levels and microscopic findings of hepatic apoptosis/necrosis were observed in both species. Finally, the dog appeared to be much more sensitive to IAP antagonism with comparable adverse effects being observed at doses that were 10- to 15-fold lower than those in the rat. Our current work investigating concentration-effect relationships between GDC-0152 and MCP-1 suggests that dogs are > 30-fold more sensitive to IAP inhibition compared with rats based on the higher estimated value of E and n for the dog (Table 4), and the resulting steeper relationship between GDC-0152 concentration and MCP-1 response in dogs compared with rats (Fig. 4).

Overall, the preclinical toxicology findings were consistent with the described single-agent mechanism of action and the associated NF-κB activation and subsequent upregulation of

FIG. 4. Plot of GDC-0152 concentration vs. % increase in MCP-1 forma-tion rate for rat and dog.

TABLE 5PK Parameter Estimates for GDC-0152 in Human (n = 36)

PK parametersPopulation mean

(%SE)Interindividual variance

(%SE)

kpc (h−1) 1.06 (7.25) —

kcp (h−1) 2.52 (5.48) 0.00263 (56.7)

kpc2 (h−1) 0.159 (9.43) —

kcp2 (h−1) 0.228 (16.9) 0.129 (35.8)

Vc (l) 8.49 (9.69) 0.151 (53.5)ke (h

−1) 5.26 (6.73) 0.0573 (45.7)Residual variability Proportional error (σprop) 0.0813 (16.9) —

TABLE 4TD Parameter Estimates from MCP-1 Population TK/PD Model for Rat and Dog

TD parameters

Rat (n = 16) Dog (n = 40)

Population mean (%SE) Interindividual variance (%SE) Population mean (%SE) Interindividual variance (%SE)

K (h−1) 0.727 (7.45) 0.0511 (33.4) 0.584 (3.59) 0.0256 (37.4)E (µM−n) 1.05 (37.0) 1.86 (13.2) 30.5 (34.9) 4.57 (28.0)kin (pg*mL−1h−1) 14.2 (12.4) 0.122 (45.3) 125 (26.1) 2.21 (27.1)Ro (pg/ml) 128 (4.19) 0.0224 (43.4) 132 (16.2) 0.899 (24.6)n 1.26 (18.7) 0.533 (1.84) 3.02 (11.1) 0.183 (41.5)Residual variability Proportional error (σprop) 0.245 (6.24) — 0.334 (6.75) —

210 WONG ET AL.

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

TNF-α. Cancer cells have been shown to frequently overex-press IAP proteins where they serve as regulators of cancer cell survival and are often indicators of poor prognosis (Vucic and Fairbrother, 2007). An increase in local TNF-α in cancer tumor cells overexpressing IAP proteins would be essential for any single-agent antitumor effects of IAP antagonists. However, normal cells are also subject to the effects of IAP antagon-ism and widespread NF-κB activation can result in adverse events due to a TNF-α driven inflammatory response. In clin-ical trials, TNF-α driven adverse events ranging from hypo-tension, thrombocytopenia, leukopenia, neurotoxicity, fever,

and nausea/vomiting to more serious events such as hypoten-sive shock proved to be limiting in the exploration of recom-binant TNF-α as an anticancer agent (Roberts et  al., 2011). From a therapeutic standpoint, for IAP antagonists to succeed as anticancer agents, local upregulation of TNF-α must occur in cancer cells before significant systemic TNF-α upregula-tion occurs. Systemic MCP-1, a chemokine that plays a critical role in recruitment of monocytes during the body’s response to inflammation or infection (Yadav et al., 2010), was identified as the most sensitive downstream marker of TNF-α upregulation and NF-κB activation in toxicology studies with GDC-0152

FIG. 5. Simulations of % change of MCP-1 vs. time using human pharmacokinetics and rat (A) or dog (B) TD parameters at iv doses of 0.76, 1.06, and 1.48 mg/kg. These doses represent the doses in the last three cohorts of reported data from the GDC-0152 clinical trial.

FIG. 6. The maximum % change in MCP-1 observed in each cohort of the clinical trial in cancer patients. Maximum % change was observed from 4.5 to 18.5 h postdose in cancer patient samples.

TK/TD ANALYSIS OF MCP-1 211

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

(Erickson et  al., 2011). In our studies, MCP-1 served as a marker of widespread systemic TNF-α upregulation.

TK-TD modeling offers a tool that improves the translata-bility of preclinical safety findings to humans. It provides a more mechanistic and robust quantitative assessment of con-centration/toxicity relationships beyond more traditional evalu-ations of dose or exposure (i.e., AUC)/toxicity relationships and also provides a means to account for species differences in pharmacokinetics (Wong et  al., 2012a,b). Our work with MCP-1 shows significant species differences in sensitivity to the TNF-α driven systemic inflammatory response as a con-sequence of IAP antagonism. The difference in the steepness of the relationship between GDC-0152 plasma concentration and the percent increase in MCP-1 formation rate between rat and dog illustrates a dramatic species difference in the systemic inflammatory response to IAP inhibition, with the dog being clearly more sensitive (Fig.  4). More importantly, we utilize existing clinical data to assess the risk of IAP antagonism in humans. Based upon simulations of MCP-1 response using human pharmacokinetics from the phase 1a clinical trial, the sensitivity of the TNF-α driven inflammatory response exhib-ited by dogs does not appear to be duplicated in humans. The simulations suggest up to ~850–2400% increase in MCP-1 lev-els in humans at the two highest doses tested (i.e., 1.06 and 1.48 mg/kg) if human responded similarly to dogs (Fig. 5). The observed maximum changes in MCP-1 levels in cancer patients at these doses were considerably less and were considered within the normal variability observed in patients.

Our findings with GDC-0152 are consistent with literature describing the dog’s sensitivity to endotoxin-induced shock, a process that is also mediated by TNF-α and results in inflam-matory and acute-phase protein responses and hepatic injury. The dog is ~100-fold more sensitive to endotoxin compared with the rat (Hamano et al., 2002). Compared with monkeys, dogs exhibit markedly enhanced effects of endotoxin on portal and systemic arterial pressures, which are rapidly increased and decreased, respectively (Brobmann et al., 1970). This unique sensitivity to endotoxin observed in dogs has been partially attributed to larger increases in Kupffer cell-mediated release of cytokines (IL-1β and IL-6) in dogs following endotoxin stimulation (Hamano et  al., 2002). Also of note, dogs have large amounts of smooth muscle in sphincter-like configura-tions around the hepatic vein which contract in response to vasoactive cytokines, resulting in congestive hepatopathy and ischemic liver injury (Aydinli and Bayraktar, 2007; Legare and Lautt, 1987). This unique feature of dog anatomy may in part explain the observed vasodynamic differences between canine and primate species. In terms of relevance to humans, responses to endotoxin-induced shock in monkeys appear to be more rep-resentative of humans (Cavanagh et al., 1970).

Our conclusions that dogs are much more sensitive than humans to IAP antagonism are consistent with emerging clinical data on IAP antagonists, LCL161, HGS1029, and TL32711 (Graham et al., 2011; Infante et al., 2010; Sikic et al., 2011). All

compounds have been reported to be well tolerated thus far in clinical trials. TL32711 and LCL161 have reached anticipated target efficacious exposures. Administration of LCL161 and HGS1029 has resulted in cytokine and chemokine upregulation with minimal adverse effects and no reported signs of a severe TNF-α driven systemic inflammatory response. Taken together, these data to date suggest that the systemic inflammatory response to IAP inhibition in humans is more similar to that observed in rats rather than in dogs. In rats, TNF-α driven systemic inflammatory response (as assessed by MCP-1 changes) appears to increase relatively gradually with increasing concentrations of IAP antagonist compared with the dog. Therefore, the expectation is that therapeutic concentrations of IAP antagonists can be achieved in humans prior to observations of dose-limiting adverse events associated with upregulation of TNF-α and other inflammatory cytokines and chemokines. In summary, our work on GDC-0152 presents for the first time, direct evidence based on MCP-1 changes in preclinical species and humans that dogs are more sensitive than humans and rats to IAP antagonism. Finally, our work presents a means of utilizing existing preclinical and clinical data and TK/TD modeling in order to provide a more quantitative assessment of the safety risk of IAP antagonists to humans and properly context the adverse findings observed in dogs.

SUPPLEMENTARY DATA

Supplementary data are available online at http://toxsci.oxfordjournals.org/.

FUNDING

This research was funded by entirely by Genentech.

ACKNOWLEDGMENTS

The authors thank the patients and their families that were involved in the clinical trial. The authors also thank members of the DMPK and Safety Assessment departments that were involved in the generation of the data in this manuscript. S. Gail Eckhardt received fellowship funding and consulting fees from Genentech. Andrew Wagner received research funding and consulting fees from Genentech.

REFERENCES

Aydinli, M., and Bayraktar, Y. (2007). Budd-Chiari syndrome: Etiology, patho-genesis and diagnosis. World J. Gastroenterol. 13, 2693–2696.

Bauer, R. J., and Guzy, S. (2004). Monte Carlo parametric expectation maxi-mization (MC-PEM) method for analyzing population pharmacokinetic/pharmacodynamic (PK/PD) data. In Advanced Methods of Pharmacokinetic and Pharmacodynamic System Analysis (D. Z. D’Argenio, Ed.), pp. 135–163. Kluwer, Boston, MA.

212 WONG ET AL.

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from

Brobmann, G. F., Ulano, H. B., Hinshaw, L. B., and Jacobson, E. D. (1970). Mesenteric vascular responses to endotoxin in the monkey and dog. Am. J. Physiol. 219, 1464–1467.

Cavanagh, D., Rao, P. S., Sutton, D. M., Bhagat, B. D., and Bachmann, F. (1970). Pathophysiology of endotoxin shock in the primate. Am. J. Obstet. Gynecol. 108, 705–722.

D’Argenio, D., and Schumitzky, A. (1997). ADAPT II User’s Guide: Pharmacokinetic/Pharmacodynamic System Analysis Software. Biomedical Simulations Resources, Los Angeles, CA.

Davies, B., and Morris, T. (1993). Physiological parameters in laboratory ani-mals and humans. Pharm. Res. 10, 1093–1095.

DeGeorge, J. J., Ahn, C. H., Andrews, P. A., Brower, M. E., Giorgio, D. W., Goheer, M. A., Lee-Ham, D. Y., McGuinn, W. D., Schmidt, W., Sun, C. J., et al. (1998). Regulatory considerations for preclinical development of anti-cancer drugs. Cancer Chemother. Pharmacol. 41, 173–185.

Dueber, E. C., Schoeffler, A. J., Lingel, A., Elliott, J. M., Fedorova, A. V., Giannetti, A. M., Zobel, K., Maurer, B., Varfolomeev, E., Wu, P., et al. (2011). Antagonists induce a conformational change in cIAP1 that promotes autoubiquitination. Science 334, 376–380.

Erickson, R., Tarrant, J., Cain, G., Dybdal, N., Lewin-Koh, S. C., Wong, H., Blackwood, E., West, K., Almeida, D., Amemiya, K., et  al. (2011). Toxicology profile of small-molecule IAP Antagonist GDC-0152 is associ-ated to TNF-alpha pharmacology. American College of Toxicology Annual Meeting Abstract.

Flygare, J. A., Beresini, M., Budha, N., Chan, H., Chan, I. T., Cheeti, S., Cohen, F., Deshayes, K., Doerner, K., Eckhardt, S. G., et al. (2012). Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J. Med. Chem. 55, 4101–4113.

Flygare, J. A., and Fairbrother, W. J. (2010). Small-molecule pan-IAP antago-nists: A patent review. Expert Opin. Ther. Pat. 20, 251–267.

Graham, M. A., Mitsuuchi, Y., Burns, J., Chunduru, S., Benetatos, C., McKinlay, M., Weng, D., Wick, M. J., Tolcher, A. W., Papadopoulos, K., et al. (2011). Phase 1 PK/PD analysis of the Smac-mimetic TL32711 dem-onstrates potent and sustained cIAP1 suppression in patient PBMCs and tumor biopsies. AACR-NCI-EORTC International Conference Abstract, Nov 2011, San Francisco, CA.

Gilbaldi, M., and Perrier, D. (1982). Pharmacokinetics, 2nd ed. Marcel Dekker Inc., New York, NY.

Hamano, T., Tong, V., Mutai, M., Hayashi, M., and Tanaka, E. (2002). Kupffer cell-mediated cytotoxicity induced by lipopolysaccharide 0111:B4 is greater in dogs than in rats and monkeys. J. Toxicol. Sci. 27, 1–9.

ICH. (2010). Guidance for Industry: S9 Nonclinical Evaluation for Anticancer Pharmaceuticals.

Infante, J. R., Dees, E. C., Burris, H. A., Zawei, L., Sager, J. A., Stevenson, C., Clarke, K., Dhuria, S., Porter, D., Sen, S. K., et al. (2010). A phase I study

of LCL161, an oral IAP inhibitor, in patients with advanced cancer. AACR 101st Annual Meeting 2010 Abstract, April 2010, Washington, DC.

Legare, D. J., and Lautt, W. W. (1987). Hepatic venous resistance site in the dog: Localization and validation of intrahepatic pressure measurements. Can. J. Physiol. Pharmacol. 65, 352–359.

Ndubaku, C., Varfolomeev, E., Wang, L., Zobel, K., Lau, K., Elliott, L. O., Maurer, B., Fedorova, A. V., Dynek, J. N., Koehler, M., et  al. (2009). Antagonism of c-IAP and XIAP proteins is required for efficient induction of cell death by small-molecule IAP antagonists. ACS Chem. Biol. 4, 557–566.

Roberts, N. J., Zhou, S., Diaz, L. A., Jr, and Holdhoff, M. (2011). Systemic use of tumor necrosis factor alpha as an anticancer agent. Oncotarget 2, 739–751.

Sikic, B. I., Eckhardt, S. G., Gallant, G., Burris, H. A., Camidge, D. R., Colevas, A. D., Jones, S. F., Messersmith, W. A., Wakelee, H. A., Li, H., et al. (2011). Safety, pharmacokinetics (PK), and pharmacodynamics (PD) of HGS1029, an inhibitor of apoptosis protein (IAP) inhibitor, in patients (Pts) with advanced solid tumors: Results of a phase I study. J. Clin. Oncol. 29, 3008 (Abstract).

Varfolomeev, E., Alicke, B., Elliott, J. M., Zobel, K., West, K., Wong, H., Scheer, J. M., Ashkenazi, A., Gould, S. E., Fairbrother, W. J., et al. (2009). X chromosome-linked inhibitor of apoptosis regulates cell death induction by proapoptotic receptor agonists. J. Biol. Chem. 284, 34553–34560.

Varfolomeev, E., Blankenship, J. W., Wayson, S. M., Fedorova, A. V., Kayagaki, N., Garg, P., Zobel, K., Dynek, J. N., Elliott, L. O., Wallweber, H. J., et al. (2007). IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis. Cell 131, 669–681.

Varfolomeev, E., and Vucic, D. (2011). Inhibitor of apoptosis proteins: Fascinating biology leads to attractive tumor therapeutic targets. Future Oncol. 7, 633–648.

Vince, J. E., Wong, W. W., Khan, N., Feltham, R., Chau, D., Ahmed, A. U., Benetatos, C. A., Chunduru, S. K., Condon, S. M., McKinlay, M., et  al. (2007). IAP antagonists target cIAP1 to induce TNFalpha-dependent apop-tosis. Cell 131, 682–693.

Vucic, D., and Fairbrother, W. J. (2007). The inhibitor of apoptosis proteins as therapeutic targets in cancer. Clin. Cancer Res. 13, 5995–6000.

Wong, H., Choo, E. F., Alicke, B., Ding, X., La, H., McNamara, E., Theil, F. P., Tibbitts, J., Friedman, L. S., Hop, C. E., et al. (2012a). Antitumor activity of targeted and cytotoxic agents in murine subcutaneous tumor models cor-relates with clinical response. Clin. Cancer Res. 18, 3846–3855.

Wong, H., Vernillet, L., Peterson, A., Ware, J. A., Lee, L., Martini, J. F., Yu, P., Li, C., Del Rosario, G., Choo, E. F., et al. (2012b). Bridging the gap between preclinical and clinical studies using pharmacokinetic-pharmacodynamic modeling: An analysis of GDC-0973, a MEK inhibitor. Clin. Cancer Res. 18, 3090–3099.

Yadav, A., Saini, V., and Arora, S. (2010). MCP-1: Chemoattractant with a role beyond immunity: A review. Clin. Chim. Acta 411, 1570–1579.

TK/TD ANALYSIS OF MCP-1 213

by guest on October 17, 2012

http://toxsci.oxfordjournals.org/D

ownloaded from


Recommended