SlideShare a Scribd company logo
Original article
A rat pharmacokinetic/pharmacodynamic model for assessment of
lipopolysaccharide-induced tumor necrosis factor-alpha production
Qin Wang a,⁎, Yuhua Zhang b,1
, J. Perry Hall b
, Lih-Ling Lin b
, Uma Raut a
, Nevena Mollova a,2
,
Neal Green c
, John Cuozzo b,3
, Shannon Chesley a
, Xin Xu a
, Jeremy I. Levin c
, Vikram S. Patel a
a
Department of Discovery Pharmacokinetics, Wyeth Research, 1 Burtt Road, Andover, MA 01810, USA
b
Department of Inflammation, 200 Cambridge Park Drive, Cambridge, MA 02140, USA
c
Department of Chemical and Screening Science, 200 Cambridge Park Drive, Cambridge, MA 02140, USA
Received 25 October 2006; accepted 14 January 2007
Abstract
Introduction: Tumor necrosis factor-alpha (TNFα) participates in many inflammatory processes. TNFα modulators show beneficial effects for the
treatment of many diseases including rheumatoid arthritis. The purpose of this study was to validate a rat pharmacokinetic/pharmacodynamic (PK/PD)
model for rapid assessment of drug candidates that intended to interrupt TNFα synthesis or release. Methods: Rats received intravenous (IV) or oral
administrations of test article or dose vehicle, followed by LPS challenge. Plasma levels of test article and TNFα were determined. The areas under the
concentration–time curves (AUCdrug and AUCTNFα) were calculated. The overall percentage of inhibition on TNFα release in vivo was calculated by
comparing AUCTNFα of the test article treated group against that for the vehicle control group. Results: The dosing vehicles tested in this study did not
increase plasma TNFα level. At IV dose of up to 100 μg/kg, LPS did not alter the pharmacokinetics of the compound tested. Using a selective TNFα
converting enzyme (TACE) inhibitor as model compound, this PK/PD model demonstrated its ability to correlate plasma test article concentration with
its biological activity of lowering the LPS-induced TNFα plasma levels in vivo. Discussion: A rat PK/PD model for evaluation of the effect of drug
candidates on LPS-induced TNFα synthesis and/or release has been investigated. This model provides integrated information on pharmacokinetics and
in vivo potency of the test articles.
© 2007 Elsevier Inc. All rights reserved.
Keywords: PK/PD model for TNFα release; Rat LPS model
1. Introduction
Recent advances in our understanding of the pathogenesis of
rheumatoid arthritis (RA) reveal that tumor necrosis factor-alpha
(TNFα) plays a pivotal role in progression of the disease. Three
recombinant proteins that neutralize TNFα effectively alleviate
the clinical symptoms of RA. Etanercept, a fusion protein of the
soluble form of the human TNFα receptor p75 and the Fc portion
of human IgG1, binds to circulating TNFα with high affinity and
thereby, blocks the biological functions of TNFα. Infliximab, a
chimeric monoclonal antibody that contains a murine TNFα
binding variable region within human immunoglobulin G1
(IgG1), prevents the interaction of TNFα with its cellular
receptor signaling targets (Nahar, Shojania, Marra, Alamgir, &
Anis, 2003). Adalimumab, a fully humanized anti-TNFα anti-
body, works by a similar mechanism (Olsen & Stein, 2004;
Shanahan & St. Clair, 2002; Braun & Sieper, 2004). Based upon
the success of these biopharmaceutical therapies, it is believed
that reducing TNFα production or release can curtail or even halt
joint damage and the progression of RA (Dayer, 2004), and
perhaps, ameliorate other diseases in which TNFα plays a role
(Stokes & Kremer, 2003). There have been many attempts to
develop new medications that modulate TNFα production or
release, especially orally available small synthetic molecules.
Tumor necrosis factor-alpha converting enzyme (TACE, Black,
2002; Conway et al., 2001; Skotnicki, DiGrandi, & Levin, 2003)
Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
www.elsevier.com/locate/jpharmtox
⁎ Corresponding author. Tel.: +1 978 247 1364; fax: +1 978 247 2842.
E-mail address: qwang@wyeth.com (Q. Wang).
1
Current address: P&G Pharmaceuticals, Cincinnati, OH 45040, USA.
2
Current address: CV therapeutics, Palo Alto, CA 94304, USA.
3
Current address: Praecis Pharmaceuticals, Waltham, MA 02451, USA.
1056-8719/$ - see front matter © 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.vascn.2007.02.001
and p38 mitogen activated protein kinase (p38 MAPK,
Schieven, 2005) are among the most studied therapeutic targets
in this regard.
The major sources of TNFα are monocytes and macro-
phages. Other cell types that produce TNFα include T
lymphocytes, NK cells, dendritic cells, mast cells, endothelial
cells, smooth muscle cells, osteoblasts, and astrocytes (Aggar-
wal, Samanta, & Feldmann, 2001). Avariety of noxious stimuli,
ranging from physical to chemical and immunological, can
induce TNFα production and release, both in vitro and in vivo.
For example, after administration of bacterial Lipopolysaccha-
ride (LPS) to animals, the blood levels of TNFα increase
rapidly. This, in turn, causes the production of pathophysiolog-
ical changes similar to those observed under inflammatory
conditions (Feldmann & Maini, 2001). Although the relevance
of LPS-stimulated pathways to the pathogenesis of inflammatory
diseases such as RA remains to be determined, it is nevertheless a
common practice to evaluate drug candidates by monitoring the
inhibition of LPS-induced TNFα production (Beck et al., 2002;
Mclay et al., 2001; Zhang, Xu et al., 2004). From drug screening
points of view, it is necessary to link test articles in vitro potency
with its in vivo activities. The information obtained from such
relationship helps to design dosing regimens for chronic disease
models such as collagen-induced arthritis (CIA).
In this report, using a selective TACE inhibitor, TMI-2
(Zhang, Hegen et al., 2004), we have validated a rat PK/PD
model for rapid screening and ranking of drug candidates that
are intended to inhibit TNFα synthesis or release for the
treatment of inflammatory diseases, such as RA.
2. Materials and methods
2.1. Chemicals and test materials
Unless specified, all chemicals, reagents and excipients of
dose formulations, were obtained from Aldrich-Sigma (St
Louis, MO) and used as received. The dose of LPS (E. coli
O111: B4, Lot # B59097, Aldrich-Sigma) was prepared in a
phosphate buffered saline solution (PBS, pH 7.2 to 7.4).
Deionized water, saline and PBS were freshly prepared in the
laboratory. Test article, TMI-2, was synthesized at medicinal
chemistry laboratory of Wyeth Research. Saline and an organic
dose vehicle (N-methyl-pyrrolidone: polyethylene glycol 400:
propylene glycol, 10:40:50, v/v/v) were used as IV dose ve-
hicles for hydrophilic and lipophilic compounds, respectively.
Oral dose formulation was an aqueous suspension containing
2% polysorbate 80 and 0.5% methylcellulose. The dose volume
for the test article was 1 mL/kg for IV and 5 mL/kg for oral
administration. All doses, LPS and test articles, were freshly
prepared on the day of the study.
2.2. Procedures for animal studies
The study was performed at Wyeth Research Laboratory
(Andover, MA) under the supervision of the Institutional
Animal Care and Use Committee (IACUC). The animals used
in the study were female Lewis rats (Charles River Laboratories,
Wilmington, MA) and weighed between 200 to 350 g. Rats had
jugular vein catheters surgically implanted prior to their arrival
at Wyeth. Briefly, the animals, divided into various treatment
and control groups (n=4 per group), received single doses of
test article or dosing vehicle via either tail vein injection or oral
gavage. The TMI-2 dose were 5 mg/kg for IVand 10 mg/kg for
oral administration, respectively. Blood samples of approxi-
mately 300 μL were collected at various time points through
jugular vein catheter into pediatric plasma separator tubes
containing K2-EDTA. The total blood lost per animal was less
than 15% of rat blood volume. Immediately after the 15 min
time point blood collection, an IV dose of LPS (100 μg/kg) in
PBS was administered via the jugular vein catheter followed by
approximately 200 to 250 μL of blank saline. Blood samples
collection continued after LPS challenge. Plasma samples
were stored at −80 °C until assay for TNFα and test article
concentrations.
2.3. Determination of plasma TNFα concentration
The detailed procedures of in vitro IC50 determination of
TACE inhibitors in rat and human whole blood were described
in a previous report (Zhang, Hegen et al., 2004). Briefly, freshly
collected whole blood was fortified with a test article of various
concentrations, followed by stimulation with LPS (100 ng/mL)
and incubation at 37 °C for about 3 h under gentle rotation. The
blood samples were then centrifuged at 1500 rpm for 15 min.
The plasma samples were collected and frozen at −80 °C until
analysis. The plasma TNFα levels, from both in vitro and in
vivo studies, were determined by a commercial ELISA kit
(BioSource International, Camarillo, CA).
2.4. Liquid Chromatography–Mass Spectrometry (LC–MS/MS)
assay for plasma test article levels
The separation of test articles from plasma samples was
performed on a Perkin Elmer Series 200 HPLC system (Perkin
Elmer, Norwalk, CT) using a XTerra MS C18 column
(2.1×20 mm, 2.5 μm; Waters, Milford, MA). The mobile
phase consisted of Solvent A (0.1% HCOOH in H2O) and
Solvent B (0.1% HCOOH in acetonitrile). The detection of test
article was performed on a PE SCIEX API-3000 triple quad-
rupole mass spectrometer (Applied Biosystems, Concord,
Ontario, L4K4V8) using a TurboIon Spray source. Briefly, an
aliquot of 50 μL plasma was precipitated with 100 μL
68 Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
acetonitrile containing 500 ng/mL of internal standard, a
compound with similar chemical structure as that for the test
article, in a 0.5 mL 96-well plate. The mixtures were vortexed
and centrifuged at 5700 rpm for 10 min. Supernatants were
directly subjected to LC–MS/MS system for analysis. The
gradient for HPLC elution was isocratic at 0% of Solvent B for
1 min, followed by a linear increase to 100% of Solvent B over
3 min. The column was allowed to equilibrate with 0% Solvent B
for 2 min before the next injection. The flow rate was 0.2 mL/
min and the injection volume was 10 μL. The plasma standard
curves were generated by plotting peak area ratio of test article
and internal standard against nominal concentrations. The limit
of quantitation for TMI-2 in rat plasma was 1 ng/mL.
2.5. Pharmacokinetic–pharmacodynamic (PK/PD) analysis
The pharmacokinetic parameters were computed using
WinNonlin (version 4.1, Pharsight, Mountain View, CA) via
non-compartmental analysis approaches. The estimation of the
area under the plasma concentration versus time curve (AUC) of
TMI-2 (AUCTMI-2) was based upon log–linear trapezoidal rule.
However, the AUC of TNFα (AUCTNFα) was computed by
linear trapezoidal method. The overall percentage of reduction
on TNFα release was calculated by the AUCTNFα ratio of TMI-
2 treated versus dose vehicle treated control groups.
For PK/PD modeling, a first order two compartment model
for IV or a first order no lag time one compartment model for
oral administration of the test article, and a Hill equation
(inhibitory effect Emax sigmoid model) were best fitted for the
plasma TMI-2 and TNFα concentration–time profiles (model
106 in WinNonlin), respectively. No statistical analysis was
conducted.
3. Results
3.1. Effect of dose vehicles on plasma TNFα concentration
Given that many chemicals stimulate TNFα release, it was
important to examine and select dose vehicles that did not
produce such effects. For the two IV dose formulations, saline
and in particular, the organic vehicle (N-methyl-pyrrolidone:
polyethylene glycol 400: propylene glycol, 10:40:50, v:v:v),
there were no increases in plasma TNFα concentrations. The
average baseline TNFα levels were below 0.6 ng/mL. After
IV bolus injection of LPS at 100 μg/kg, plasma TNFα started
to rise by approximately 30 min and reached its peak
concentration at approximately 90 min. The plasma TNFα
level then underwent rapid decline to baseline by about 3 to
4 h post LPS administration (Fig. 1). The average Cmax of
TNFα ranged between 8 and 10 ng/m with mean plasma
AUCTNFα value of approximately 14–18 ng×hr/mL. As ex-
pected, the aqueous oral dose vehicle did not increase plasma
TNFα levels.
3.2. Effect of LPS on the absorption and disposition of test
article
LPS is a toxic agent that induces a wide array of biological,
biochemical, and immunological reactions (Ray, 1999). There-
fore, it was necessary to validate the effects of concomitant LPS
Fig. 1. Organic dose vehicle of 1 mL/kg (N-methyl-pyrrolidone: polyethylene
glycol 400: propylene glycol, 10:40:50, v:v:v, IV, open circle) did not cause
observable changes in plasma TNFα after 100 μg/kg LPS challenge.
Fig. 2. After IV (2a, 5 mg/kg) and PO (2b, 10 mg/kg) administrations, the
plasma concentration–time profiles of TMI-2 were not altered by concomitant
administration of 100 μg/kg LPS dose.
69Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
administration on the pharmacokinetics of test articles in
animals. Following IVor oral administration to rats, the plasma
concentration–time profiles of TMI-2 were essentially super-
imposable to those obtained from studies in which LPS was not
dosed (Fig. 2a and b).
3.3. Effect of TNFα modulator on plasma TNFα levels upon
LPS challenge
The plasma concentration–time profiles of TNFα, after IV
or oral administration of TMI-2 followed by LPS challenge,
were plotted in Fig. 3a and b. The overall reduction of TNFα
(AUCTNFα) was 69% after IV treatment at AUCTMI-2 of
2266 ng×h/mL and 81% after oral treatment at AUCTMI-2 of
1450 ng×h/mL. The lesser overall reduction of TNFα
release on higher AUCTMI-2 for IV treatment group could
be due to the timing of the LPS challenge, which occurred at
15 min post TMI-2 dose administration. By then, plasma
concentration had dropped significantly so that a large
portion of the plasma exposure of TMI-2 did not participate
in the inhibition of LPS induced TNFα production–release
process. Since TACE is at the last step of TNFα release and is a
membrane protein of blood cells, test article in plasma directly
interacts with the target. An inhibitory effect sigmoid Emax
model (Eq. (1)) best fitted the PK/PD link model.
E ¼ E0 þ Emax−E0ð Þ Â
Cg
plasma
Cg
plasma þ ECg
50
ð1Þ
where E is the percent of reduction of TNFα, at any given
time, assuming complete prevention of TNFα release (Emax)
can be achieved if plasma test article level (Cplasma) is
sufficiently high. E0 is the baseline from vehicle treated
group. EC50 is 50% reduction in TNFα release (Fig. 4). Here,
γ is the curve shape factor for the Hill equation (Nigrovic &
Amann, 2002).
Based on the curve fitting, the in vivo EC50 for TMI-2 was
approximately 303 ng/mL and 71 ng/mL after IV and oral dose
administration, respectively. The results were in agreement with
in vitro EC50 of 70–100 ng/mL for TMI-2 from rat whole blood
assay.
4. Discussion
LPS-induced TNFα production and release can be simplified
as a series of events that starts with LPS binding to LPS binding
protein (LPB), and that complex then binds to CD14 which
initiates a signaling cascade that culminates in expression of the
tnfα gene, processing of the TNFα precursor protein, insertion
of the TNFα precursor in the cell membrane, cleavage of the
precursor, and finally, release of the active form of TNFα, a
17 kDa soluble fragment, into the extra-cellular spaces
(Anderson, Phillips, Stoecklin, & Kedersha, 2004; Gracie,
Leung, & McInnes, 2002). Alterations in the concentration of
TNFα in fresh blood (or cell culture media) can be expected if
the synthesis or release of TNFα is disrupted in animals (or cell
culture models) during the response to LPS challenge. By
Fig. 3. Plasma concentration–time profiles of TNFα after IV (3a, 5 mg/kg) and
oral (3b, 10 mg/kg) administrations of TMI-2 followed by 100 μg/ml LPS
stimulation.
Fig. 4. An inhibitory effect sigmoid Emax PK/PD link model was best fitted for
the reduction in plasma TNFα levels in the presence of TMI-2. The chart on the
upper right shows the plasma concentration–time profiles of TMI-2 after IV or
oral administration.
70 Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
choosing an appropriate PK/PD model, one can establish an in
vitro–in vivo correlation for drug candidates that are intended to
modulate this cascade.
At the early phase of drug discovery, drug candidates may
not possess needed potency, selectivity and/or pharmacokinetic
properties. By directly introducing test articles to animals via IV
injection at relatively higher doses, formulated in organic
vehicles if necessary, a PK/PD study can serve as a proof-of-
concept for target validation and a quick assessment of in vitro–
in vivo correlation. Unlike mouse model (Gozzi et al., 1999),
rats provide a quantity of blood volume per animal that is suffi-
cient for complete assessment of plasma (or serum) concentra-
tion–time profiles of both test article and TNFα. This engenders
less interference in the data from inter-animal variations.
In this report, using a selective TACE inhibitor as model
compound, we have studied a rat PK/PD model for validation
for drug candidates that are intended to modulate TNFα levels.
Acknowledgements
We thank Jessica Doherty, Cindy Clark, Amy Ignatowicz,
Terrie Cunliffe-Beamer, and Glen Pedneault for their excellent
technical supports.
References
Aggarwal, B. B., Samanta, A., & Feldmann, M. (2001). TNFα. In J. J.
Oppenheim & M. Feldmann (Eds.), Cytokine reference: A compendium of
cytokines and other mediators of host defense (1st ed.). San Diego:
Academic Press.
Anderson, P., Phillips, K., Stoecklin, G., & Kedersha, N. (2004). Post-
transcriptional regulation of proinflammatory proteins. Journal of Leukocyte
Biology, 76(1), 42−47.
Beck, G., Bottomley, G., Bradshaw, D., Brewster, M., Broadhurst, M., Devos,
R., et al. (2002). (E)-2(R)-[1(S)-(Hydroxycarbamoyl)-4-phenyl-3-butenyl]-
2′-isobutyl-2′-(methanesulfonyl)-4-methylvalerohydrazide (Ro 32–7315), a
selective and orally active inhibitor of tumor necrosis factor-α convertase.
Journal of Pharmacology and Experimental Therapeutics, 302, 390−396.
Black, R. A. (2002). Tumor necrosis factor-alpha converting enzyme. Interna-
tional Journal of Biochemistry & Cell Biology, 34(1), 1−5.
Braun, J., & Sieper, J. (2004). Biological therapies in the spondyloarthritides—
The current state. Rheumatology, 43(9), 1072−1084.
Conway, J. G., Andrews, R. C., Beaudet, B., Bickett, D. M., Boncek, V., Brodie,
T. A., et al. (2001). Inhibition of tumor necrosis factor-α (TNF-α)
production and arthritis in the rat by GW3333, a dual inhibitor of TNFα
converting enzyme and matrix metalloproteinases. Journal of Pharmacol-
ogy and Experimental Therapeutics, 298, 900−908.
Dayer, J. M. (2004). The process of identifying and understanding cytokines:
From basic studies to treating rheumatic diseases. Best Practice & Research.
Clinical Rheumatology, 18(1), 31−45.
Feldmann, M., & Maini, R. N. (2001). Anti-TNFα therapy of rheumatoid
arthritis: What have we learned? Annual Review of Immunology, 19,
163−196.
Gozzi, P., Pahlman, I., Palmer, L., Gronberg, A., & Persson, S. (1999).
Pharmacokinetic-pharmacodynamic modeling of the immunomodulating
agent susalimod and experimentally induced tumor necrosis factor-alpha
levels in the mouse. Journal of Pharmacology & Experimental Therapeu-
tics, 291(1), 199−203 (Oct).
Gracie, J. A., Leung, B. P., & McInnes, I. B. (2002). Novel pathways that
regulate tumor necrosis factor-alpha production in rheumatoid arthritis.
Current Opinion in Rheumatology, 14(3), 270−275.
Mclay, L. M., Halley, F., Souness, J. E., McKenna, J., Benning, V., Birrell, M.,
et al. (2001). The discovery of RPR 200765A, a p38 MAP kinase inhibitor
displaying a good oral anti-arthritic efficacy. Bioorganic & Medicinal
Chemistry, 9(2), 537−554.
Nahar, I. K., Shojania, K., Marra, C. A., Alamgir, A. H., & Anis, A. H. (2003).
Inflixmab treatment of rheumatoid arthritis and crohn's disease. Annals of
Pharmacotherapy, 37, 1256−1265.
Nigrovic, V., & Amann, A. (2002). Physiologic–pharmacologic interpretation of
the constants in the hill equation for neuromuscular block: A hypothesis.
Journal of Pharmacokinetics and Pharmacodanamics, 29(2), 189−206.
Olsen, N. J., & Stein, C. M. (2004). New drug for rheumatoid arthritis. New
England Journal of Medicine, 350, 2167−2179.
Ray, P. K. (1999). Stress genes and species survival. Molecular and Cellular
Biochemistry, 196(1–2), 117−123.
Shanahan, J. C., & St. Clair, E. W. (2002). Short analytical review tumor
necrosis factor-α blockage: A novel therapy for rheumatic disease. Clinical
Immunobiology, 103, 231−242.
Schieven, G. L. (2005). The biology of p38 kinase: A central role in
inflammation. Current Topics in Medicinal Chemistry, 5(10), 921−928.
Skotnicki, J. S., DiGrandi, M. J., & Levin, J. I. (2003). Design strategies for the
identification of MMP-13 and Tace inhibitors. Current Opinion in Drug
Discovery & Development, 6(5), 742−759.
Stokes, D. G., & Kremer, J. M. (2003). Potential of tumor necrosis factor
neutralization strategies in rheumatologic disorders other than rheumatoid
arthritis. Seminars in Arthritis and Rheumatism, 33(1), 1−18.
Zhang, Y., Xu, J., Levin, J., Hegen, M., Li, G., Robertshaw, H., et al. (2004).
Identification and characterization of 4-[[4-(2-butynyloxy)phenyl]sulfonyl]-
N-hydroxy-2,2-dimethyl-(3S)thiomorpholinecarboxamide (TMI-1), a novel
dual tumor necrosis factor-alpha-converting enzyme/matrix metalloprotease
inhibitor for the treatment of rheumatoid arthritis. Journal of Pharmacology
and Experimental Therapeutics, 309(1), 348−355.
Zhang, Y., Hegen, M., Xu, J., Keith, J., Jin, G., Du, X., et al. (2004).
Characterization of (2R, 3S)-2-([4-(2-butynyloxy)phenyl]sulfonyl amino)-
N,3-dihydroxybutanamide, a potent and selective inhibitor of TNF-α
converting enzyme. International Immunopharmacology, 4(14), 1845−1857.
71Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71

More Related Content

What's hot

Mutagencity and its types ppt
Mutagencity and its types pptMutagencity and its types ppt
Mutagencity and its types ppt
Sravanthi Shetty
 
Development and Validation of a Two-Site Immunoradiometric assay for Glypican...
Development and Validation of a Two-Site Immunoradiometric assay for Glypican...Development and Validation of a Two-Site Immunoradiometric assay for Glypican...
Development and Validation of a Two-Site Immunoradiometric assay for Glypican...
Premier Publishers
 
Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...
Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...
Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...
International Journal of Engineering Inventions www.ijeijournal.com
 
Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...
Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...
Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...
Brandi VanAlphen
 
Acetogenins In Vivo re: McLaughlin
Acetogenins In Vivo re: McLaughlinAcetogenins In Vivo re: McLaughlin
Acetogenins In Vivo re: McLaughlin
Ben Rockefeller
 
articulo
articulo articulo
articulo
Monserrath96
 
3. RAAS- MEWARI- IJBMR-2013
3. RAAS- MEWARI- IJBMR-20133. RAAS- MEWARI- IJBMR-2013
3. RAAS- MEWARI- IJBMR-2013
Pulakes Purkait
 
bioequivalence
bioequivalencebioequivalence
bioequivalence
Giovanny Leon
 
Structurally characterized arabinogalactan from Anoectochilus formosanus as a...
Structurally characterized arabinogalactan from Anoectochilus formosanus as a...Structurally characterized arabinogalactan from Anoectochilus formosanus as a...
Structurally characterized arabinogalactan from Anoectochilus formosanus as a...
Cây thuốc Việt
 
6. endo reg 2018_52_02_abdelaleem_59-68
6. endo reg 2018_52_02_abdelaleem_59-686. endo reg 2018_52_02_abdelaleem_59-68
6. endo reg 2018_52_02_abdelaleem_59-68
Minia university, Faculty of Medicine
 
Kit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile DuctKit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile Duct
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
Cytotoxicity Screening Simarouba Tulae
Cytotoxicity Screening Simarouba TulaeCytotoxicity Screening Simarouba Tulae
Cytotoxicity Screening Simarouba Tulae
University of Puerto Rico at Cayey
 
Hofstetter PON1 meeting 2015 KOmice corrected
Hofstetter PON1 meeting 2015 KOmice correctedHofstetter PON1 meeting 2015 KOmice corrected
Hofstetter PON1 meeting 2015 KOmice corrected
Catherine A. Hofstetter
 
Liver enzymes in normal and sickle cell
Liver enzymes in normal and sickle cellLiver enzymes in normal and sickle cell
Liver enzymes in normal and sickle cell
Alexander Decker
 
CrossGen-Merck manuscript
CrossGen-Merck manuscriptCrossGen-Merck manuscript
CrossGen-Merck manuscript
Kush Sharma
 
Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...
Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...
Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...
iosrjce
 
Liver ischemia/reperfusion injury, a setting in which the functional mass is ...
Liver ischemia/reperfusion injury, a setting in which the functional mass is ...Liver ischemia/reperfusion injury, a setting in which the functional mass is ...
Liver ischemia/reperfusion injury, a setting in which the functional mass is ...
Prof. Hesham N. Mustafa
 
Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...
Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...
Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
In vivo studies of wound healing and hepatoprotective agents
In vivo studies of wound healing and hepatoprotective agentsIn vivo studies of wound healing and hepatoprotective agents
In vivo studies of wound healing and hepatoprotective agents
Adarsh Patil
 
Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...
Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...
Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...
pharmaindexing
 

What's hot (20)

Mutagencity and its types ppt
Mutagencity and its types pptMutagencity and its types ppt
Mutagencity and its types ppt
 
Development and Validation of a Two-Site Immunoradiometric assay for Glypican...
Development and Validation of a Two-Site Immunoradiometric assay for Glypican...Development and Validation of a Two-Site Immunoradiometric assay for Glypican...
Development and Validation of a Two-Site Immunoradiometric assay for Glypican...
 
Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...
Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...
Protective Effect of Salacia Oblanga and Quercetin on Cyclophosphamide-Induce...
 
Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...
Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...
Quantitation of Capsaicin Levels in Hot Peppers by Gas Chromatography/Mass Sp...
 
Acetogenins In Vivo re: McLaughlin
Acetogenins In Vivo re: McLaughlinAcetogenins In Vivo re: McLaughlin
Acetogenins In Vivo re: McLaughlin
 
articulo
articulo articulo
articulo
 
3. RAAS- MEWARI- IJBMR-2013
3. RAAS- MEWARI- IJBMR-20133. RAAS- MEWARI- IJBMR-2013
3. RAAS- MEWARI- IJBMR-2013
 
bioequivalence
bioequivalencebioequivalence
bioequivalence
 
Structurally characterized arabinogalactan from Anoectochilus formosanus as a...
Structurally characterized arabinogalactan from Anoectochilus formosanus as a...Structurally characterized arabinogalactan from Anoectochilus formosanus as a...
Structurally characterized arabinogalactan from Anoectochilus formosanus as a...
 
6. endo reg 2018_52_02_abdelaleem_59-68
6. endo reg 2018_52_02_abdelaleem_59-686. endo reg 2018_52_02_abdelaleem_59-68
6. endo reg 2018_52_02_abdelaleem_59-68
 
Kit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile DuctKit-Positive Cells in the Murine Common Bile Duct
Kit-Positive Cells in the Murine Common Bile Duct
 
Cytotoxicity Screening Simarouba Tulae
Cytotoxicity Screening Simarouba TulaeCytotoxicity Screening Simarouba Tulae
Cytotoxicity Screening Simarouba Tulae
 
Hofstetter PON1 meeting 2015 KOmice corrected
Hofstetter PON1 meeting 2015 KOmice correctedHofstetter PON1 meeting 2015 KOmice corrected
Hofstetter PON1 meeting 2015 KOmice corrected
 
Liver enzymes in normal and sickle cell
Liver enzymes in normal and sickle cellLiver enzymes in normal and sickle cell
Liver enzymes in normal and sickle cell
 
CrossGen-Merck manuscript
CrossGen-Merck manuscriptCrossGen-Merck manuscript
CrossGen-Merck manuscript
 
Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...
Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...
Neuroendocrine Response to Stress In Tame Versus Untame Grasscutters (Thryono...
 
Liver ischemia/reperfusion injury, a setting in which the functional mass is ...
Liver ischemia/reperfusion injury, a setting in which the functional mass is ...Liver ischemia/reperfusion injury, a setting in which the functional mass is ...
Liver ischemia/reperfusion injury, a setting in which the functional mass is ...
 
Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...
Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...
Synergy Assessment of Hypoxia and Lipopolysaccharide on Autophagy in Dendriti...
 
In vivo studies of wound healing and hepatoprotective agents
In vivo studies of wound healing and hepatoprotective agentsIn vivo studies of wound healing and hepatoprotective agents
In vivo studies of wound healing and hepatoprotective agents
 
Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...
Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...
Pharmacological evaluation of hepatoprotective activity of ethanolic extract ...
 

Viewers also liked

Gail donegan's html cv
Gail donegan's html cvGail donegan's html cv
Gail donegan's html cv
Gail Donegan
 
2016 Annual General & Special Meeting of Shareholders
2016 Annual General & Special Meeting of Shareholders2016 Annual General & Special Meeting of Shareholders
2016 Annual General & Special Meeting of Shareholders
Critical Outcome Technologies Inc.
 
Immunobiology and new challenges in drug development
Immunobiology and new challenges in drug developmentImmunobiology and new challenges in drug development
Immunobiology and new challenges in drug development
Dr Kurt Sales
 
Resume and Full CV for RJW_Oct 2015
Resume and Full CV for RJW_Oct 2015Resume and Full CV for RJW_Oct 2015
Resume and Full CV for RJW_Oct 2015
Raymond Winquist
 
Sol padis dec10-llp-2013
Sol padis dec10-llp-2013Sol padis dec10-llp-2013
Sol padis dec10-llp-2013
Stanford University
 
Deecher D Cv 3.5.09
Deecher D Cv 3.5.09Deecher D Cv 3.5.09
Deecher D Cv 3.5.09
guest20d1ac
 
Hui Zhang CV 2016
Hui Zhang CV 2016Hui Zhang CV 2016
Hui Zhang CV 2016
Hui Zhang
 
CV 5 17 15
CV 5 17 15CV 5 17 15
CV 5 17 15
Mike Lauer
 
Dr. David Banji - CV
Dr. David Banji - CVDr. David Banji - CV
Dr. David Banji - CV
Prof. Dr. David Banji
 

Viewers also liked (9)

Gail donegan's html cv
Gail donegan's html cvGail donegan's html cv
Gail donegan's html cv
 
2016 Annual General & Special Meeting of Shareholders
2016 Annual General & Special Meeting of Shareholders2016 Annual General & Special Meeting of Shareholders
2016 Annual General & Special Meeting of Shareholders
 
Immunobiology and new challenges in drug development
Immunobiology and new challenges in drug developmentImmunobiology and new challenges in drug development
Immunobiology and new challenges in drug development
 
Resume and Full CV for RJW_Oct 2015
Resume and Full CV for RJW_Oct 2015Resume and Full CV for RJW_Oct 2015
Resume and Full CV for RJW_Oct 2015
 
Sol padis dec10-llp-2013
Sol padis dec10-llp-2013Sol padis dec10-llp-2013
Sol padis dec10-llp-2013
 
Deecher D Cv 3.5.09
Deecher D Cv 3.5.09Deecher D Cv 3.5.09
Deecher D Cv 3.5.09
 
Hui Zhang CV 2016
Hui Zhang CV 2016Hui Zhang CV 2016
Hui Zhang CV 2016
 
CV 5 17 15
CV 5 17 15CV 5 17 15
CV 5 17 15
 
Dr. David Banji - CV
Dr. David Banji - CVDr. David Banji - CV
Dr. David Banji - CV
 

Similar to a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysaccharide-induced-tumor-necrosis-factor-alpha-production

Mol Pharmacol-2003-Jones-471-7
Mol Pharmacol-2003-Jones-471-7Mol Pharmacol-2003-Jones-471-7
Mol Pharmacol-2003-Jones-471-7
Suzanne Holden
 
Jianying Xiao CV 2016
Jianying Xiao CV 2016  Jianying Xiao CV 2016
Jianying Xiao CV 2016
Xiao Jianying
 
00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...
00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...
00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...
Minia university, Faculty of Medicine
 
BRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docx
BRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docxBRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docx
BRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docx
hartrobert670
 
2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)
2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)
2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)
Ji-Youn Yeo
 
journalism journals
journalism journalsjournalism journals
journalism journals
chaitanya451336
 
research on journaling
research on journalingresearch on journaling
research on journaling
chaitanya451336
 
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
[14796813   journal of molecular endocrinology] identification of tacc1, nov,...[14796813   journal of molecular endocrinology] identification of tacc1, nov,...
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
University of Zambia, School of Pharmacy, Lusaka, Zambia
 
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
[14796813   journal of molecular endocrinology] identification of tacc1, nov,...[14796813   journal of molecular endocrinology] identification of tacc1, nov,...
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
University of Zambia, School of Pharmacy, Lusaka, Zambia
 
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
[14796813   journal of molecular endocrinology] identification of tacc1, nov,...[14796813   journal of molecular endocrinology] identification of tacc1, nov,...
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
University of Zambia, School of Pharmacy, Lusaka, Zambia
 
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Sean Ekins
 
I0342048053
I0342048053I0342048053
I0342048053
iosrphr_editor
 
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in RatsProtective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
Fertility Week - April 2014 Sample Issue
Fertility Week - April 2014 Sample IssueFertility Week - April 2014 Sample Issue
Fertility Week - April 2014 Sample Issue
Varun Swamy
 
sclabas2005
sclabas2005sclabas2005
sclabas2005
Christian Schmidt
 
A common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organsA common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organs
Kevin Jaglinski
 
Lan Gấm 1 - Nghiên cứu khoa học
Lan Gấm 1 - Nghiên cứu khoa họcLan Gấm 1 - Nghiên cứu khoa học
Lan Gấm 1 - Nghiên cứu khoa học
Cây thuốc Việt
 
Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...
Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...
Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...
science journals
 
Fall2014_ResearchPoster_CodyHeiser
Fall2014_ResearchPoster_CodyHeiserFall2014_ResearchPoster_CodyHeiser
Fall2014_ResearchPoster_CodyHeiser
Cody Heiser
 
Bilal CAPR-14-0231.full
Bilal CAPR-14-0231.fullBilal CAPR-14-0231.full
Bilal CAPR-14-0231.full
Bilal Bin Hafeez, M.Sc, Ph.D
 

Similar to a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysaccharide-induced-tumor-necrosis-factor-alpha-production (20)

Mol Pharmacol-2003-Jones-471-7
Mol Pharmacol-2003-Jones-471-7Mol Pharmacol-2003-Jones-471-7
Mol Pharmacol-2003-Jones-471-7
 
Jianying Xiao CV 2016
Jianying Xiao CV 2016  Jianying Xiao CV 2016
Jianying Xiao CV 2016
 
00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...
00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...
00.toxicological assessment-of-venlafaxine-acute-and-subchronic-toxicity-stud...
 
BRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docx
BRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docxBRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docx
BRIEF OBSERVATIONAll-cause Mortality Associated withTNF-a .docx
 
2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)
2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)
2014-Yeo-A Multiplex Two-Color Real-Time PCR Method(1)
 
journalism journals
journalism journalsjournalism journals
journalism journals
 
research on journaling
research on journalingresearch on journaling
research on journaling
 
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
[14796813   journal of molecular endocrinology] identification of tacc1, nov,...[14796813   journal of molecular endocrinology] identification of tacc1, nov,...
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
 
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
[14796813   journal of molecular endocrinology] identification of tacc1, nov,...[14796813   journal of molecular endocrinology] identification of tacc1, nov,...
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
 
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
[14796813   journal of molecular endocrinology] identification of tacc1, nov,...[14796813   journal of molecular endocrinology] identification of tacc1, nov,...
[14796813 journal of molecular endocrinology] identification of tacc1, nov,...
 
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
Using Machine Learning Models Based on Phenotypic Data to Discover New Molecu...
 
I0342048053
I0342048053I0342048053
I0342048053
 
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in RatsProtective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
 
Fertility Week - April 2014 Sample Issue
Fertility Week - April 2014 Sample IssueFertility Week - April 2014 Sample Issue
Fertility Week - April 2014 Sample Issue
 
sclabas2005
sclabas2005sclabas2005
sclabas2005
 
A common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organsA common rejection module (CRM) for acute rejection across multiple organs
A common rejection module (CRM) for acute rejection across multiple organs
 
Lan Gấm 1 - Nghiên cứu khoa học
Lan Gấm 1 - Nghiên cứu khoa họcLan Gấm 1 - Nghiên cứu khoa học
Lan Gấm 1 - Nghiên cứu khoa học
 
Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...
Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...
Geriatric Care at a Time of Accelerated Aging in the World Population and Eme...
 
Fall2014_ResearchPoster_CodyHeiser
Fall2014_ResearchPoster_CodyHeiserFall2014_ResearchPoster_CodyHeiser
Fall2014_ResearchPoster_CodyHeiser
 
Bilal CAPR-14-0231.full
Bilal CAPR-14-0231.fullBilal CAPR-14-0231.full
Bilal CAPR-14-0231.full
 

a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysaccharide-induced-tumor-necrosis-factor-alpha-production

  • 1. Original article A rat pharmacokinetic/pharmacodynamic model for assessment of lipopolysaccharide-induced tumor necrosis factor-alpha production Qin Wang a,⁎, Yuhua Zhang b,1 , J. Perry Hall b , Lih-Ling Lin b , Uma Raut a , Nevena Mollova a,2 , Neal Green c , John Cuozzo b,3 , Shannon Chesley a , Xin Xu a , Jeremy I. Levin c , Vikram S. Patel a a Department of Discovery Pharmacokinetics, Wyeth Research, 1 Burtt Road, Andover, MA 01810, USA b Department of Inflammation, 200 Cambridge Park Drive, Cambridge, MA 02140, USA c Department of Chemical and Screening Science, 200 Cambridge Park Drive, Cambridge, MA 02140, USA Received 25 October 2006; accepted 14 January 2007 Abstract Introduction: Tumor necrosis factor-alpha (TNFα) participates in many inflammatory processes. TNFα modulators show beneficial effects for the treatment of many diseases including rheumatoid arthritis. The purpose of this study was to validate a rat pharmacokinetic/pharmacodynamic (PK/PD) model for rapid assessment of drug candidates that intended to interrupt TNFα synthesis or release. Methods: Rats received intravenous (IV) or oral administrations of test article or dose vehicle, followed by LPS challenge. Plasma levels of test article and TNFα were determined. The areas under the concentration–time curves (AUCdrug and AUCTNFα) were calculated. The overall percentage of inhibition on TNFα release in vivo was calculated by comparing AUCTNFα of the test article treated group against that for the vehicle control group. Results: The dosing vehicles tested in this study did not increase plasma TNFα level. At IV dose of up to 100 μg/kg, LPS did not alter the pharmacokinetics of the compound tested. Using a selective TNFα converting enzyme (TACE) inhibitor as model compound, this PK/PD model demonstrated its ability to correlate plasma test article concentration with its biological activity of lowering the LPS-induced TNFα plasma levels in vivo. Discussion: A rat PK/PD model for evaluation of the effect of drug candidates on LPS-induced TNFα synthesis and/or release has been investigated. This model provides integrated information on pharmacokinetics and in vivo potency of the test articles. © 2007 Elsevier Inc. All rights reserved. Keywords: PK/PD model for TNFα release; Rat LPS model 1. Introduction Recent advances in our understanding of the pathogenesis of rheumatoid arthritis (RA) reveal that tumor necrosis factor-alpha (TNFα) plays a pivotal role in progression of the disease. Three recombinant proteins that neutralize TNFα effectively alleviate the clinical symptoms of RA. Etanercept, a fusion protein of the soluble form of the human TNFα receptor p75 and the Fc portion of human IgG1, binds to circulating TNFα with high affinity and thereby, blocks the biological functions of TNFα. Infliximab, a chimeric monoclonal antibody that contains a murine TNFα binding variable region within human immunoglobulin G1 (IgG1), prevents the interaction of TNFα with its cellular receptor signaling targets (Nahar, Shojania, Marra, Alamgir, & Anis, 2003). Adalimumab, a fully humanized anti-TNFα anti- body, works by a similar mechanism (Olsen & Stein, 2004; Shanahan & St. Clair, 2002; Braun & Sieper, 2004). Based upon the success of these biopharmaceutical therapies, it is believed that reducing TNFα production or release can curtail or even halt joint damage and the progression of RA (Dayer, 2004), and perhaps, ameliorate other diseases in which TNFα plays a role (Stokes & Kremer, 2003). There have been many attempts to develop new medications that modulate TNFα production or release, especially orally available small synthetic molecules. Tumor necrosis factor-alpha converting enzyme (TACE, Black, 2002; Conway et al., 2001; Skotnicki, DiGrandi, & Levin, 2003) Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71 www.elsevier.com/locate/jpharmtox ⁎ Corresponding author. Tel.: +1 978 247 1364; fax: +1 978 247 2842. E-mail address: qwang@wyeth.com (Q. Wang). 1 Current address: P&G Pharmaceuticals, Cincinnati, OH 45040, USA. 2 Current address: CV therapeutics, Palo Alto, CA 94304, USA. 3 Current address: Praecis Pharmaceuticals, Waltham, MA 02451, USA. 1056-8719/$ - see front matter © 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.vascn.2007.02.001
  • 2. and p38 mitogen activated protein kinase (p38 MAPK, Schieven, 2005) are among the most studied therapeutic targets in this regard. The major sources of TNFα are monocytes and macro- phages. Other cell types that produce TNFα include T lymphocytes, NK cells, dendritic cells, mast cells, endothelial cells, smooth muscle cells, osteoblasts, and astrocytes (Aggar- wal, Samanta, & Feldmann, 2001). Avariety of noxious stimuli, ranging from physical to chemical and immunological, can induce TNFα production and release, both in vitro and in vivo. For example, after administration of bacterial Lipopolysaccha- ride (LPS) to animals, the blood levels of TNFα increase rapidly. This, in turn, causes the production of pathophysiolog- ical changes similar to those observed under inflammatory conditions (Feldmann & Maini, 2001). Although the relevance of LPS-stimulated pathways to the pathogenesis of inflammatory diseases such as RA remains to be determined, it is nevertheless a common practice to evaluate drug candidates by monitoring the inhibition of LPS-induced TNFα production (Beck et al., 2002; Mclay et al., 2001; Zhang, Xu et al., 2004). From drug screening points of view, it is necessary to link test articles in vitro potency with its in vivo activities. The information obtained from such relationship helps to design dosing regimens for chronic disease models such as collagen-induced arthritis (CIA). In this report, using a selective TACE inhibitor, TMI-2 (Zhang, Hegen et al., 2004), we have validated a rat PK/PD model for rapid screening and ranking of drug candidates that are intended to inhibit TNFα synthesis or release for the treatment of inflammatory diseases, such as RA. 2. Materials and methods 2.1. Chemicals and test materials Unless specified, all chemicals, reagents and excipients of dose formulations, were obtained from Aldrich-Sigma (St Louis, MO) and used as received. The dose of LPS (E. coli O111: B4, Lot # B59097, Aldrich-Sigma) was prepared in a phosphate buffered saline solution (PBS, pH 7.2 to 7.4). Deionized water, saline and PBS were freshly prepared in the laboratory. Test article, TMI-2, was synthesized at medicinal chemistry laboratory of Wyeth Research. Saline and an organic dose vehicle (N-methyl-pyrrolidone: polyethylene glycol 400: propylene glycol, 10:40:50, v/v/v) were used as IV dose ve- hicles for hydrophilic and lipophilic compounds, respectively. Oral dose formulation was an aqueous suspension containing 2% polysorbate 80 and 0.5% methylcellulose. The dose volume for the test article was 1 mL/kg for IV and 5 mL/kg for oral administration. All doses, LPS and test articles, were freshly prepared on the day of the study. 2.2. Procedures for animal studies The study was performed at Wyeth Research Laboratory (Andover, MA) under the supervision of the Institutional Animal Care and Use Committee (IACUC). The animals used in the study were female Lewis rats (Charles River Laboratories, Wilmington, MA) and weighed between 200 to 350 g. Rats had jugular vein catheters surgically implanted prior to their arrival at Wyeth. Briefly, the animals, divided into various treatment and control groups (n=4 per group), received single doses of test article or dosing vehicle via either tail vein injection or oral gavage. The TMI-2 dose were 5 mg/kg for IVand 10 mg/kg for oral administration, respectively. Blood samples of approxi- mately 300 μL were collected at various time points through jugular vein catheter into pediatric plasma separator tubes containing K2-EDTA. The total blood lost per animal was less than 15% of rat blood volume. Immediately after the 15 min time point blood collection, an IV dose of LPS (100 μg/kg) in PBS was administered via the jugular vein catheter followed by approximately 200 to 250 μL of blank saline. Blood samples collection continued after LPS challenge. Plasma samples were stored at −80 °C until assay for TNFα and test article concentrations. 2.3. Determination of plasma TNFα concentration The detailed procedures of in vitro IC50 determination of TACE inhibitors in rat and human whole blood were described in a previous report (Zhang, Hegen et al., 2004). Briefly, freshly collected whole blood was fortified with a test article of various concentrations, followed by stimulation with LPS (100 ng/mL) and incubation at 37 °C for about 3 h under gentle rotation. The blood samples were then centrifuged at 1500 rpm for 15 min. The plasma samples were collected and frozen at −80 °C until analysis. The plasma TNFα levels, from both in vitro and in vivo studies, were determined by a commercial ELISA kit (BioSource International, Camarillo, CA). 2.4. Liquid Chromatography–Mass Spectrometry (LC–MS/MS) assay for plasma test article levels The separation of test articles from plasma samples was performed on a Perkin Elmer Series 200 HPLC system (Perkin Elmer, Norwalk, CT) using a XTerra MS C18 column (2.1×20 mm, 2.5 μm; Waters, Milford, MA). The mobile phase consisted of Solvent A (0.1% HCOOH in H2O) and Solvent B (0.1% HCOOH in acetonitrile). The detection of test article was performed on a PE SCIEX API-3000 triple quad- rupole mass spectrometer (Applied Biosystems, Concord, Ontario, L4K4V8) using a TurboIon Spray source. Briefly, an aliquot of 50 μL plasma was precipitated with 100 μL 68 Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
  • 3. acetonitrile containing 500 ng/mL of internal standard, a compound with similar chemical structure as that for the test article, in a 0.5 mL 96-well plate. The mixtures were vortexed and centrifuged at 5700 rpm for 10 min. Supernatants were directly subjected to LC–MS/MS system for analysis. The gradient for HPLC elution was isocratic at 0% of Solvent B for 1 min, followed by a linear increase to 100% of Solvent B over 3 min. The column was allowed to equilibrate with 0% Solvent B for 2 min before the next injection. The flow rate was 0.2 mL/ min and the injection volume was 10 μL. The plasma standard curves were generated by plotting peak area ratio of test article and internal standard against nominal concentrations. The limit of quantitation for TMI-2 in rat plasma was 1 ng/mL. 2.5. Pharmacokinetic–pharmacodynamic (PK/PD) analysis The pharmacokinetic parameters were computed using WinNonlin (version 4.1, Pharsight, Mountain View, CA) via non-compartmental analysis approaches. The estimation of the area under the plasma concentration versus time curve (AUC) of TMI-2 (AUCTMI-2) was based upon log–linear trapezoidal rule. However, the AUC of TNFα (AUCTNFα) was computed by linear trapezoidal method. The overall percentage of reduction on TNFα release was calculated by the AUCTNFα ratio of TMI- 2 treated versus dose vehicle treated control groups. For PK/PD modeling, a first order two compartment model for IV or a first order no lag time one compartment model for oral administration of the test article, and a Hill equation (inhibitory effect Emax sigmoid model) were best fitted for the plasma TMI-2 and TNFα concentration–time profiles (model 106 in WinNonlin), respectively. No statistical analysis was conducted. 3. Results 3.1. Effect of dose vehicles on plasma TNFα concentration Given that many chemicals stimulate TNFα release, it was important to examine and select dose vehicles that did not produce such effects. For the two IV dose formulations, saline and in particular, the organic vehicle (N-methyl-pyrrolidone: polyethylene glycol 400: propylene glycol, 10:40:50, v:v:v), there were no increases in plasma TNFα concentrations. The average baseline TNFα levels were below 0.6 ng/mL. After IV bolus injection of LPS at 100 μg/kg, plasma TNFα started to rise by approximately 30 min and reached its peak concentration at approximately 90 min. The plasma TNFα level then underwent rapid decline to baseline by about 3 to 4 h post LPS administration (Fig. 1). The average Cmax of TNFα ranged between 8 and 10 ng/m with mean plasma AUCTNFα value of approximately 14–18 ng×hr/mL. As ex- pected, the aqueous oral dose vehicle did not increase plasma TNFα levels. 3.2. Effect of LPS on the absorption and disposition of test article LPS is a toxic agent that induces a wide array of biological, biochemical, and immunological reactions (Ray, 1999). There- fore, it was necessary to validate the effects of concomitant LPS Fig. 1. Organic dose vehicle of 1 mL/kg (N-methyl-pyrrolidone: polyethylene glycol 400: propylene glycol, 10:40:50, v:v:v, IV, open circle) did not cause observable changes in plasma TNFα after 100 μg/kg LPS challenge. Fig. 2. After IV (2a, 5 mg/kg) and PO (2b, 10 mg/kg) administrations, the plasma concentration–time profiles of TMI-2 were not altered by concomitant administration of 100 μg/kg LPS dose. 69Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
  • 4. administration on the pharmacokinetics of test articles in animals. Following IVor oral administration to rats, the plasma concentration–time profiles of TMI-2 were essentially super- imposable to those obtained from studies in which LPS was not dosed (Fig. 2a and b). 3.3. Effect of TNFα modulator on plasma TNFα levels upon LPS challenge The plasma concentration–time profiles of TNFα, after IV or oral administration of TMI-2 followed by LPS challenge, were plotted in Fig. 3a and b. The overall reduction of TNFα (AUCTNFα) was 69% after IV treatment at AUCTMI-2 of 2266 ng×h/mL and 81% after oral treatment at AUCTMI-2 of 1450 ng×h/mL. The lesser overall reduction of TNFα release on higher AUCTMI-2 for IV treatment group could be due to the timing of the LPS challenge, which occurred at 15 min post TMI-2 dose administration. By then, plasma concentration had dropped significantly so that a large portion of the plasma exposure of TMI-2 did not participate in the inhibition of LPS induced TNFα production–release process. Since TACE is at the last step of TNFα release and is a membrane protein of blood cells, test article in plasma directly interacts with the target. An inhibitory effect sigmoid Emax model (Eq. (1)) best fitted the PK/PD link model. E ¼ E0 þ Emax−E0ð Þ Â Cg plasma Cg plasma þ ECg 50 ð1Þ where E is the percent of reduction of TNFα, at any given time, assuming complete prevention of TNFα release (Emax) can be achieved if plasma test article level (Cplasma) is sufficiently high. E0 is the baseline from vehicle treated group. EC50 is 50% reduction in TNFα release (Fig. 4). Here, γ is the curve shape factor for the Hill equation (Nigrovic & Amann, 2002). Based on the curve fitting, the in vivo EC50 for TMI-2 was approximately 303 ng/mL and 71 ng/mL after IV and oral dose administration, respectively. The results were in agreement with in vitro EC50 of 70–100 ng/mL for TMI-2 from rat whole blood assay. 4. Discussion LPS-induced TNFα production and release can be simplified as a series of events that starts with LPS binding to LPS binding protein (LPB), and that complex then binds to CD14 which initiates a signaling cascade that culminates in expression of the tnfα gene, processing of the TNFα precursor protein, insertion of the TNFα precursor in the cell membrane, cleavage of the precursor, and finally, release of the active form of TNFα, a 17 kDa soluble fragment, into the extra-cellular spaces (Anderson, Phillips, Stoecklin, & Kedersha, 2004; Gracie, Leung, & McInnes, 2002). Alterations in the concentration of TNFα in fresh blood (or cell culture media) can be expected if the synthesis or release of TNFα is disrupted in animals (or cell culture models) during the response to LPS challenge. By Fig. 3. Plasma concentration–time profiles of TNFα after IV (3a, 5 mg/kg) and oral (3b, 10 mg/kg) administrations of TMI-2 followed by 100 μg/ml LPS stimulation. Fig. 4. An inhibitory effect sigmoid Emax PK/PD link model was best fitted for the reduction in plasma TNFα levels in the presence of TMI-2. The chart on the upper right shows the plasma concentration–time profiles of TMI-2 after IV or oral administration. 70 Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71
  • 5. choosing an appropriate PK/PD model, one can establish an in vitro–in vivo correlation for drug candidates that are intended to modulate this cascade. At the early phase of drug discovery, drug candidates may not possess needed potency, selectivity and/or pharmacokinetic properties. By directly introducing test articles to animals via IV injection at relatively higher doses, formulated in organic vehicles if necessary, a PK/PD study can serve as a proof-of- concept for target validation and a quick assessment of in vitro– in vivo correlation. Unlike mouse model (Gozzi et al., 1999), rats provide a quantity of blood volume per animal that is suffi- cient for complete assessment of plasma (or serum) concentra- tion–time profiles of both test article and TNFα. This engenders less interference in the data from inter-animal variations. In this report, using a selective TACE inhibitor as model compound, we have studied a rat PK/PD model for validation for drug candidates that are intended to modulate TNFα levels. Acknowledgements We thank Jessica Doherty, Cindy Clark, Amy Ignatowicz, Terrie Cunliffe-Beamer, and Glen Pedneault for their excellent technical supports. References Aggarwal, B. B., Samanta, A., & Feldmann, M. (2001). TNFα. In J. J. Oppenheim & M. Feldmann (Eds.), Cytokine reference: A compendium of cytokines and other mediators of host defense (1st ed.). San Diego: Academic Press. Anderson, P., Phillips, K., Stoecklin, G., & Kedersha, N. (2004). Post- transcriptional regulation of proinflammatory proteins. Journal of Leukocyte Biology, 76(1), 42−47. Beck, G., Bottomley, G., Bradshaw, D., Brewster, M., Broadhurst, M., Devos, R., et al. (2002). (E)-2(R)-[1(S)-(Hydroxycarbamoyl)-4-phenyl-3-butenyl]- 2′-isobutyl-2′-(methanesulfonyl)-4-methylvalerohydrazide (Ro 32–7315), a selective and orally active inhibitor of tumor necrosis factor-α convertase. Journal of Pharmacology and Experimental Therapeutics, 302, 390−396. Black, R. A. (2002). Tumor necrosis factor-alpha converting enzyme. Interna- tional Journal of Biochemistry & Cell Biology, 34(1), 1−5. Braun, J., & Sieper, J. (2004). Biological therapies in the spondyloarthritides— The current state. Rheumatology, 43(9), 1072−1084. Conway, J. G., Andrews, R. C., Beaudet, B., Bickett, D. M., Boncek, V., Brodie, T. A., et al. (2001). Inhibition of tumor necrosis factor-α (TNF-α) production and arthritis in the rat by GW3333, a dual inhibitor of TNFα converting enzyme and matrix metalloproteinases. Journal of Pharmacol- ogy and Experimental Therapeutics, 298, 900−908. Dayer, J. M. (2004). The process of identifying and understanding cytokines: From basic studies to treating rheumatic diseases. Best Practice & Research. Clinical Rheumatology, 18(1), 31−45. Feldmann, M., & Maini, R. N. (2001). Anti-TNFα therapy of rheumatoid arthritis: What have we learned? Annual Review of Immunology, 19, 163−196. Gozzi, P., Pahlman, I., Palmer, L., Gronberg, A., & Persson, S. (1999). Pharmacokinetic-pharmacodynamic modeling of the immunomodulating agent susalimod and experimentally induced tumor necrosis factor-alpha levels in the mouse. Journal of Pharmacology & Experimental Therapeu- tics, 291(1), 199−203 (Oct). Gracie, J. A., Leung, B. P., & McInnes, I. B. (2002). Novel pathways that regulate tumor necrosis factor-alpha production in rheumatoid arthritis. Current Opinion in Rheumatology, 14(3), 270−275. Mclay, L. M., Halley, F., Souness, J. E., McKenna, J., Benning, V., Birrell, M., et al. (2001). The discovery of RPR 200765A, a p38 MAP kinase inhibitor displaying a good oral anti-arthritic efficacy. Bioorganic & Medicinal Chemistry, 9(2), 537−554. Nahar, I. K., Shojania, K., Marra, C. A., Alamgir, A. H., & Anis, A. H. (2003). Inflixmab treatment of rheumatoid arthritis and crohn's disease. Annals of Pharmacotherapy, 37, 1256−1265. Nigrovic, V., & Amann, A. (2002). Physiologic–pharmacologic interpretation of the constants in the hill equation for neuromuscular block: A hypothesis. Journal of Pharmacokinetics and Pharmacodanamics, 29(2), 189−206. Olsen, N. J., & Stein, C. M. (2004). New drug for rheumatoid arthritis. New England Journal of Medicine, 350, 2167−2179. Ray, P. K. (1999). Stress genes and species survival. Molecular and Cellular Biochemistry, 196(1–2), 117−123. Shanahan, J. C., & St. Clair, E. W. (2002). Short analytical review tumor necrosis factor-α blockage: A novel therapy for rheumatic disease. Clinical Immunobiology, 103, 231−242. Schieven, G. L. (2005). The biology of p38 kinase: A central role in inflammation. Current Topics in Medicinal Chemistry, 5(10), 921−928. Skotnicki, J. S., DiGrandi, M. J., & Levin, J. I. (2003). Design strategies for the identification of MMP-13 and Tace inhibitors. Current Opinion in Drug Discovery & Development, 6(5), 742−759. Stokes, D. G., & Kremer, J. M. (2003). Potential of tumor necrosis factor neutralization strategies in rheumatologic disorders other than rheumatoid arthritis. Seminars in Arthritis and Rheumatism, 33(1), 1−18. Zhang, Y., Xu, J., Levin, J., Hegen, M., Li, G., Robertshaw, H., et al. (2004). Identification and characterization of 4-[[4-(2-butynyloxy)phenyl]sulfonyl]- N-hydroxy-2,2-dimethyl-(3S)thiomorpholinecarboxamide (TMI-1), a novel dual tumor necrosis factor-alpha-converting enzyme/matrix metalloprotease inhibitor for the treatment of rheumatoid arthritis. Journal of Pharmacology and Experimental Therapeutics, 309(1), 348−355. Zhang, Y., Hegen, M., Xu, J., Keith, J., Jin, G., Du, X., et al. (2004). Characterization of (2R, 3S)-2-([4-(2-butynyloxy)phenyl]sulfonyl amino)- N,3-dihydroxybutanamide, a potent and selective inhibitor of TNF-α converting enzyme. International Immunopharmacology, 4(14), 1845−1857. 71Q. Wang et al. / Journal of Pharmacological and Toxicological Methods 56 (2007) 67–71