Top Banner
Book of Abstracts FIFTEENTH INTERNATIONAL CONFERENCE Sunday, September 21 – Wednesday, September 24, 2008 Westfields Marriott, Chantilly, VA, USA
57
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: IRA_15th_Conference_21.-24.9.2008

Book of Abstracts

FIFTEENTH INTERNATIONAL CONFERENCE

Sunday, September 21 – Wednesday, September 24, 2008Westfields Marriott, Chantilly, VA, USA

Page 2: IRA_15th_Conference_21.-24.9.2008

LOGIN FOR THOMSON SCIENTIFIC CONGRESSREPORTS

Accessing the 15th International IRA ConferenceMeeting Summaries

As the official media partner to the InflammationResearch Association at the 15th International Confer-ence on Inflammation, Thomson Scientific will beproviding delegates with a collection of reviews featuringthe latest intelligence on investigational drugs andresearch released at the Conference. The reviews will bewritten by Thomson Scientific!s “on-site Conferencereporter” and will be accessible daily during the Confer-ence through the official Conference WEB site.Features of this Service:

• Available one week in advance of the Conference:Early birds can peruse the event preview to identifythose sessions that are a 'must-see! for those working inthe field of inflammation research.

• Daily updates : Delegates can keep abreast of thehottest news as it unfolds by accessing daily reviews ofthe latest disclosures on drug discovery and researchwith three overnight reports available on the WEB siteas the meeting progresses.

To access the meeting summaries:

Delegates can access the above collection by first loggingonto the IRAWEB site Home Page (www.inflammation-research.org), where clicking the icon marking the 15th

International IRA Conference Homepage will display amenu bar for “Scientific Congress Reports” for moreinformation and also the “Reports Login” site. Enter yourusername and the unique password to be sent to alldelegates by e-mail prior to the meeting.

If you do not have or have lost your password or you areexperiencing difficulties in logging in, please contactRichard Griffiths (860-732-5535) for further assistance.While at the meeting, please contact the IRA registrationdesk in the case of any difficulties.

Thomson Pharma/IDdb3 subscribers can also view thehistories of many of the drugs detailed, major competitorproducts, and chemical structures and patents by ac-ACHTUNGTRENNUNGcessing directly Thomson Pharma (www.thomsonpharma.com) or the IDdb (www.iddb3.com) in the usual way.

Page 3: IRA_15th_Conference_21.-24.9.2008

MISSION STATEMENT

The Inflammation Research Association is a non-profitorganization instituted to bring together scientists of alldegree and experience levels with an interest in inflam-mation research to encourage communication and discus-

sion of scientific and technological advances that can beused to develop new therapeutic agents for the widediversity of serious diseases with inflammatory processes.

LIST OF EXHIBITORS*

Active Motif Inc.Amnis CorporationBioMol International, LPBio-Quant, Inc.Birkh<user Verlag AGCalvert Laboratories, Inc.Cayman Chemical CompanyCharles RivereBioscienceEMKA TECHNOLOGIES, INC.GE HealthcareHelica Biosystems, Inc.

HistoTox Labs, Inc.MD BiosciencesMetabolon, Inc.MIR Preclinical ServicesPremier Laboratory, LLCRicerca Biosciences, LLCRules Based Medicine, Inc.Seventh Wave LaboratoriesTNO PharmaUgo Basile North America, Inc.Washington Biotechnologies, Inc.

ACKNOWLEDGEMENTS*

The Inflammation Research Association is grateful to thefollowing organizations for their financial support for the15th International Conference and other scientific meet-ings throughout the past two years:

Abbott BiosciencesAmgenArray BiopharmaBristol-Myers SquibbCelgeneCharles River LaboratoriesCombinatoRxEli LillyJohnson & JohnsonNIH-NIAID-DAITPfizer Global R&DRules Based MedicineSignal Pharmaceuticals

Sincere appreciation is extended to the following compa-nies for their additional contributions:

Abbott BiosciencesSponsorship of Symposium Novel Therapeutics:Asthma, COPD and RA

Calvert LaboratoriesRegistration, Name tags

GE HealthcarePoster Awards

HistoTox LabsCoffee break

MD Biosciences, MetabolonNewsletter

Thomson ScientificAdvertising

The ScientistAdvertising

Charles River LaboratoriesPadfolios

* As of July 1, 2008

Page 4: IRA_15th_Conference_21.-24.9.2008
Page 5: IRA_15th_Conference_21.-24.9.2008

Contents Inflammation Research

Supplement 2Vol. 57pp. S75 – S119September 2008ISSN 1023-3830

IX Welcome

X Officers and Board Members

XI Conference Organizers

XII Keynote Speaker

Symposia Abstracts

SA01 Intestinal immunomodulation by theemerging B7-like butyrophilin family

S77

SA02 The role of antigen presenting cell subsetsin regulating the balance betweenimmunity and tolerance in the intestine

S77

SA03 The Th17 lineage: development, functionand regulation

S78

SA04 Experimental breakthrough therapies inIBD

S78

SA05 Combinatorial genomics leads to rapiddiscovery of integrated pathways fortargeted therapies in IBD

S78

SA06 What do T regulatory cells (Treg) see anddo?

S80

SA07 Reshaping the immune system to inducetolerance in autoimmunity using genetherapy approaches

S80

SA08 An antigen-specific tolerance approach fortherapy of mouse models of MS and type 1diabetes

S81

SA09 Teplizumab, a humanized, Fc Receptor(FcR) non-binding anti-CD3 monoclonalantibody in recent-onset type 1 diabetes(T1DM)

S81

SA10 The surprising complexity of pain testingin the laboratory mouse

S82

SA11 Role of ASICs in muscle and jointhyperalgesia associated with inflammation

S82

SA12 Pronociceptive actions of CCL2 andCXCL12 after peripheral nerve injury

S83

SA13 Both EP4 antagonists and mPGES-1inhibitors relieve pain in rodent models ofarthritis

S83

SA14 The protein C system resides at thecrossroads of hemostasis and inflammation

S84

SA15 Biological therapeutics, T effector and Tregulatory cells in autoimmune disease

S84

SA16 Characterization and optimization ofmonoclonal antibodies for the treatmentof inflammatory diseases - case studies

S85

SA17 Biological therapeutic approach toatherosclerosis: challenges andopportunities, two case studies inexperimental disease

S85

SA18 Dissecting the IL-17 receptor: Signalingand Function

S86

SA19 The identification of a humanIL-17F/IL-17A heterodimer and thereceptor complex utilized in signaling

S86

SA20 The IL-23/IL-17 axis in arthritis S86

SA21 Stat transcription factor signaling in mouseand human Th17 polarization

S87

SA22 Inhibition of IL-4Ra : a promisingtherapeutic strategy for asthma

S88

SA23 Potential for bronchial thermoplasty toreverse airway remodeling associated withasthma

S88

SA24 BIO-11006: targeting the MARCKSprotein in COPD

S89

SA25 Anti-IL-12/IL-23 treatment for CrohnAsdisease and psoriasis

S89

SA26 Baminercept: targeting LT-b and LIGHTpathway in RA

S89

Page 6: IRA_15th_Conference_21.-24.9.2008

Mini-symposia and Poster Session Abstracts

A100 Nociceptin antagonists for the treatmentof inflammatory bowel disease

S93

A101 Design, synthesis and biological evaluationof azole derivatives of diphenyl acetic acidas anti-inflammatory agents

S93

A102 Inflammatory changes in schizophreniawith antipsychotic treatment

S93

A103 Pharmacological characterization of a newmodel of cynomolgus skin inflammationmeasured with novel methodologies

S93

A104 Biacore analysis reveals distinct receptor(a1 and a2) binding inhibitioncharacteristics of anti IL-13 antibodies

S94

A105 Micro-computed tomography (mCT) is apowerful imaging technique for evaluatingbone pathology in arthritis models

S94

A106 Soft, topical immunosuppressants derivedfrom cyclosporin A and FK-506 for thetreatment of atopic dermatitis

S94

A107 SD0006; a potent, selective, andorally-available p38 kinase inhibitor

S95

A108 A role for cathepsin K inhibitors inrheumatoid arthritis:VEL-0230 Phase I clinical trial results

S95

A109 Genomic-based high throughput screeningidentifies small molecules thatdifferentially inhibit the antiviral andimmunomodulatory effects of IFN-a

S95

A110 The induction of nuclear factor kB inlaryngeal cancer cells by humanpapillomavirus-16 oncoprotein E7 isassociated with increased inflammatorycytokines

S96

A111 Comparative effects of ananti-inflammatory blend on reducing skinirritation caused by UVB or a chemicalirritant to 1% hydrocortisone

S96

A112 Imaging an inflammatory response using19F MRI

S96

A113 A novel mouse model of Mycobacteriumtuberculosis-induced granuloma necrosis:Implications for adjuvant immunotherapytargeting TH2 cytokines in tuberculosis

S97

A114 p38 MAPK inhibitor for the treatment ofrheumatoid arthritis

S97

A115 Oral lactoferrin and glycine display in vivosynergistic anti-inflammatory activity

S97

A116 Chronic inflammation in asthma airwayremodeling

S98

A117 Effects of recombinant human growthhormone on Staphylococcus aureus sepsisin mouse

S98

A118 Suppression of C - reactive protein andlipoprotein levels in arthritic rats by novelglucosamine-analog GN1

S98

A119 Inhibitory effect of EGCG on acutepancreatitis

S99

A120 Inhibitory effect of inflammatory mediatorby Orostachys japonicus in mouseperitoneal macrophages

S99

A121 4-Aryl-5-heteroaryl-2-thio-substitutedimidazoles: Approach to p38 MAP kinaseinhibitor prodrugs

S99

A122 IL-1 drives pathogenic Th17 cells duringspontaneous arthritis in IL-1Ra-deficientmice

S100

A123 IL-17 synergy with TNF causes strikingcartilage erosion in vivo

S100

A124 Formylpeptide receptor-like-1 mediatesamyloid b-induced inflammatory responsein AlzheimerAs disease

S100

A125 Optimization of a quinolone class ofdissociated glucocorticoid mimetics

S101

A126 Inflammatory signaling processes andcardiovascular complication

S101

A127 The golgi-associated protein p115mediates the secretion of macrophagemigration inhibitory factor (MIF)

S101

A128 GABA(A)-mediated alteration ofautoimmune-mediated inflammation usingthe natural plant product, honokiol

S101

A129 Human apolipoprotein C1 transgenicmice: a unique novel model of atopicdermatitis

S102

A130 Modulation of inflammation in twohumanized mouse models of psoriasis

S102

A131 Efficacy of probiotics in TNBS-inducedcolitis

S102

A132 Predicting efficacy and side effects of thep38MAP kinase inhibitor class usingBioMAPG primary human cell-basedsystems

S102

A133 Histamine is not released in acute thermalinjury in human skin in vivo : Amicrodialysis study

S103

Page 7: IRA_15th_Conference_21.-24.9.2008

A134 Effects of a selective, potent p38 inhibitorin in vivo models of rheumatoid arthritis

S103

A135 A global gene expression profiling analysisto study the role of endothelial CD40during inflammation

S103

A136 Dipeptidyl peptidase I mediates cigarettesmoke-induced pulmonary inflammationand alveolar destruction

S104

A137 The involvement of nitric oxide in theperipheral antinociceptive effects ofopioids during chronic inflammatory pain

S104

A138 Campylobacter jejuni-induced activationof murine dendritic cells involves coopera-tive signaling through MyD88 and TRIF

S104

A139 AN2898: a novel anti-inflammatorycompound that inhibits phosphodiesterase4 and 7 enzyme activity and IL-12 andIL-23 release

S105

A140 Discovery of3-{3-(2-piperidinylethoxy)phe-nyl}-5-(1H-1,2,4-triazol-3-yl)-1H-indazole(CC-401), a potent JNK inhibitor

S105

A141 Resolvin E1 (RvE1) inhibits inflammationin acute and chronic murine models of colitis

S105

A142 The poly-anionic sugar sucroseocta-sulphate is an oral anti-rheumatic andanti-erosive metabolite of sucralfate

S105

A143 Preconditioning lymphocytes with p38MAPK inhibitors, and not accessory cells,prevents Con-A-induced lymphocyteresponses

S106

A144 Preventive role of ZH-67-2-1 inadjuvant-induced arthritis in rats

S106

A145 Design of small molecule inhibitors forinflammatory bowel diseasemechanism: inflammatory/immunemechanisms

S106

A146 L-17 signaling induces sequentialphosphorylation of C/EBPb

S107

A147 Modulatory role of 2-acetamidophenol onthe expression of CD44 cell surfacemarkers in the brain of Adjuvant InducedArthritic model (AIA) of rats

S107

A148 Protective effect of non-selective andselective COX-2-inhibitors in hypoxiastress-induced behavioral and biochemicalalterations

S108

A149 Rosiglitazone prevents hyperhomo-cysteinemia-induced myocardial remodel-ing through mast cell stabilization in rats

S108

A150 Evaluation of novel topical drug deliverysystems of colchicine in mono sodiumurate (MSU) model of gout in rats

S108

A151 Gadolinium decreases inflammationrelated to myocardial ischemia andreperfusion injury

S109

A152 The discovery of a series of novel smallmolecule macrocyclic TNF-a antagonists

S109

A153 Identification of functional roles for bothIL-17RB and IL-17RA in mediating IL-25induced activities

S109

A154 The anti-obesity effect of GyeongshinhaeGihwan T2 is associated with a decreasednitric oxide synthesis

S110

A155 Effective mechanism of herb complexprescription LAnssichegamsanA in obesity

S110

A156 Exploration of the MAP3K TAK1 as atarget for modulating inflammatory arthritis

S110

A157 Treatment with anti-CD30 ligand preventsdisease progression in murine systemiclupus erythematosus

S111

A158 Elimination of Tolmetin Ulcers byAnisodamine (ANSA)

S111

A159 Small molecule CXCR2 antagonistprevents hyperoxia-induced neutrophilaccumulation in the lungs of newborn rats

S111

A160 Synergistic induction of IL-10 by a TLRagonist and a phospho-ceramide analog ismediated by cAMP

S111

Van Arman Award Competition Abstracts

VA01 Differential modulatory effects of TLR2and TLR4 on T cell balance inexperimental arthritis: possibilities for newtherapeutic strategies

S114

VA02 The role of CC chemokine receptor 7during invasive aspergillosis

S114

VA03 Pro-inflammatory cytokines inhibitchondrogenesis of human mesenchymalstem cells through NF-kB dependentpathways

S114

VA04 Bz-423 improves GVHD-associated tissuedamage and mortality by specificallytargeting effector T cells

S115

VA05 Mapping the binding of macrophagemigration inhibitory factor (MIF) to thechemokine receptor CXCR4

S115

Author Index S117

Page 8: IRA_15th_Conference_21.-24.9.2008
Page 9: IRA_15th_Conference_21.-24.9.2008

WELCOME

September 21, 2008

Dear Colleague,

On behalf of the Inflammation Research AssociationOfficers and the Board of Directors, as well as theOrganizing Committee, we welcome you to the 15th

International Conference of the Inflammation ResearchAssociation.

In addition to excellent meeting content, we haveimplemented several format changes, including choosinga new location conveniently located just 7 miles fromDulles International Airport for easy access andproviding attendees a greater choice of topics throughconcurrent morning symposia. Of course, we will stillstress the hallmark of all IRA Conferences, the ability forclose scientific and social interactions in a relaxed, funatmosphere.

The meeting kicks off with a symposium entitled“Inflammatory Bowel Disease: From Basic Mechanismsto the Clinic” scheduled from 3-6 PM on Sunday,September 21st. The scientific program continues thatevening with a Keynote Lecture from Dr. Diane Mathis,Professor of Medicine at Harvard Medical School,speaking about “Regulation of Auto-InflammatoryResponses.” Dr. Mathis is a preeminent authority onautoimmunity and T-cell biology as it relates to inflam-matory diseases such as diabetes and rheumatoid arthritis.

Our Main Symposia on Monday the 22nd and Tuesday the23rd feature invited speakers in the following areas:

– Tolerance Induction as a Therapeutic Optionfor Autoimmunity

– Inflammation and Pain– Biologics as a Platform for Inflammation Therapies– The IL-17/IL-23 Axis in Autoimmunity

Our final symposium on Wednesday the 24th in themorning features exciting clinical data on “Novel Ther-apeutics.”

In addition to the Main Symposia, we will hold four Mini-Symposia as well as two Poster Sessions as part of theprogram. All poster presenters are automatically enteredinto a competition for the poster with the 7greatesttherapeutic potential8 that is supported by a grant fromGE Healthcare;. For scientists earlier in their careers wehave: 1) the Van Arman Scholarship Competition, whichallows five scientists to compete for cash awards whileattending the meeting with all expenses paid, and 2)networking and breakfast events for investigators new tothe inflammation arena, which will foster interaction withestablished IRA members and allow attendees to gainvaluable advice on career development.

On behalf of the IRA Officers and Board of Directors, Ilook forward to welcoming you to the 15th InternationalConference at the elegant Westfields Marriott in Chan-tilly, Virginia, and to participating in an event that fostersboth great science and social interactions.

William Selig, PhDPresidentInflammation Research Association

Page 10: IRA_15th_Conference_21.-24.9.2008

OFFICERS AND BOARD MEMBERS

2006 – 2008 Term

President William M. Selig, PhD(Magen BioSciences)

Vice-President John E. Somerville, PhD(Bristol-Myers Squibb)

Secretary Arpita Maiti, PhD(Accurate Consulting Co.)

Treasurer Andrew L. Glasebrook, PhD(Eli Lilly & Co.)

Board Members

Laurent Audoly, PhD (Merck & Co., Inc.)Brydon L. Bennett, PhD (Celgene Corp.)James L. Ellis, PhD (Surface Logix, Inc.)Richard J. Griffiths, PhD (Pfizer Inc.)Tineke Meijers, PhD (TNO Pharma)James L. Mobley, PhD (Lycera Corp.)Lisa M. Olson, PhD (Abbott Bioresearch Center)Lisa R. Schopf, PhD (Abbott Bioresearch Center)Joel E. Tocker, PhD (Amgen Inc.)Bruce E. Tomczuk, PhD (Chemnomics, LLC.)

2008 – 2010 Term

President John E. Somerville, PhD(Bristol-Myers Squibb)

Vice-President Andrew L. Glasebrook, PhD(Eli Lilly & Co.)

Secretary Joel Tocker, PhD(Amgen Inc.)

Treasurer Arpita Maiti, PhD(Accurate Consulting Co.)

Board Members

Brydon L. Bennett, PhD (Celgene Corp.)Jane Connor, PhD (MedImmune)James L. Ellis, PhD (Surface Logix, Inc.)Liwu Li, PhD (Virginia Tech)Tineke Meijers, PhD (TNO Pharma)Lisa Olson, PhD (Abbott Bioresearch Center)Lisa Schopf, PhD (Abbott Bioresearch Center)William M. Selig, PhD (Magen Biosciences)Bruce E. Tomczuk, PhD (Chemnomics, LLC.)Craig Wegner, PhD (Pfizer Inc.)

Page 11: IRA_15th_Conference_21.-24.9.2008

CONFERENCE ORGANIZERS

PlanningWilliam Selig (Magen BioSciences)John Somerville (Bristol-Myers Squibb)Arpita Maiti (Accurate Consulting Co.)Andrew Glasebrook (Eli Lilly & Co.)

Scientific ProgramJohn Somerville (Bristol-Myers Squib), ChairpersonJohn Beals (Eli Lilly & Co.)Frank Castellino (Notre Dame)Jane Connor (MedImmune)Sarah Gaffen (U. Pittsburgh)John Iacomini (Harvard)Jeffrey Mogil (McGill)Steve Nadler (Bristol-Myers Squibb)Rey Panettieri (U. Penn.)Stephan Targan (Cedars Sinai/UCLA)Joel Tocker (Amgen)Bruce Tomczuk (Chemnomics, LLC)Jo Viney (Amgen)

Mini-symposiaJim Ellis (Surface Logix), ChairpersonBill Avery (CombinatoRx)Brydon Bennett (Celgene)Brendan Canning (Johns Hopkins)Larry De Garavilla (J & J)Liwu Li (Virginia Tech)Elise Martin (Charles River Laboratories)Lisa Olson (Abbott Bioresearch Center)Lisa Schopf (Abbott Bioresearch Center)

Poster Sessions/Abstract BookEd Yurkow (J&J PRD), ChairpersonMarcia Bliven (Pfizer, retired)John Somerville (Bristol-Myers Squibb)Cindy Willis (Amgen)

Van Arman Scholarship AwardsJames Mobley (Lycera), ChairpersonLisa Olson (Abbott Bioresearch Center)Caralee Schaefer (BioSeek)Loran Killar (Pfizer, retired)Dick Dyer (Velcura Therapeutics)

GE Healthcare Poster AwardsRichard Griffiths (Pfizer Inc.)

New InvestigatorsLisa Marshall (J&J PRD), ChairpersonDavid Howat (HBS Consulting)Arpita Maiti (Accurate Consulting Co.)Lisa Schopf (Abbott Bioresearch Center)

ExhibitorsHoward Kartstein (Metabolon)

Site SelectionMarcia Bliven (Pfizer, retired)Joan Chapdelaine (Calvert Laboratories)Richard McLaughlin (Rich McLaughlin & Assoc.)Deborah Wolff (Novartis)

RegistrarJoan Chapdelaine (Calvert Laboratories)

Conference CoordinationMarcia Bliven (Pfizer, retired)Richard McLaughlin (Rich McLaughlin & Assoc.)Deborah Wolff (Novartis)

AudiovisualWilliam Selig (Magen BioSciences)

Social EventsTineke Meijers (TNO Pharma)

Fund RaisingJoel Tocker (Amgen), ChairpersonWilliam Selig (Magen BioSciences)

Advertising/ PublicityArpita Maiti (Accurate Consulting Co.), Chairperson

Thomson Drugs ProceedingsRichard Griffiths (Pfizer Inc.)

Travel BursariesWilliam Selig (Magen BioSciences)Andrew Glasebrook (Eli Lilly & Company)

Page 12: IRA_15th_Conference_21.-24.9.2008

Keynote Speaker

Diane Mathis, Ph.D.Diane Mathis obtained a B.Sc. from Wake ForestUniversity and a Ph.D. from the University of Rochester.She performed postdoctoral studies at the Laboratoire deG�n�tique Mol�culaire des Eucaryotes in Strasbourg,France and at Stanford University Medical Center. Shereturned to France at the end of 1983, establishing alaboratory at the Institut de G�n�tique et de BiologieMol�culaire et Cellulaire in Strasbourg, in conjunction

with Dr. Christophe Benoist. The lab moved to the JoslinDiabetes Center at the end of 1999. Dr. Mathis iscurrently a Professor of Medicine at Brigham andWomen8s Hospital and Harvard Medical School, and isan Associate Research Director and Head of the Sectionon Immunology and Immunogenetics at Joslin, where sheholds the William T. Young Chair in Diabetes Research.She is Director of the JDRF Center on ImmunologicalTolerance in Type-1 Diabetes at HMS, a Principle FacultyMember at the Harvard Stem Cell Institute and anAssociate Faculty Member of The Broad Institute. Dr.Mathis was elected to the U.S. Academy of Sciences in2003 and to the German Academy in 2007.

The lab works in the fields of T cell differentiation andautoimmunity, with a special emphasis on exploiting themost advanced transgenic and gene-targeting technologyto engineer new mouse models. Studies on autoimmunityexplore the immunological mechanisms of diabetes,rheumatoid arthritis and APECED, a polyglandularautoimmune disease. Major questions tackled are whatinitiates these diseases, how is their progression regulated,and what are the final effector mechanisms. In addition,modern genetic and genomic approaches are used toidentify disease-modifying genes in both human patientsand mouse models, and the application of computationaland bioinformatic strategies to these and other issues isexplored. Whole-animal imaging of inflammation and itstissular effects in diabetes and arthritis models is pursuedas part of a long-standing collaborative program.

The Mathis/Benoist laboratory has produced over 250publications and trained over 100 students and postdoc-toral fellows.

Page 13: IRA_15th_Conference_21.-24.9.2008

Symposia Abstracts

Page 14: IRA_15th_Conference_21.-24.9.2008
Page 15: IRA_15th_Conference_21.-24.9.2008

IBD from Basic Mechanisms to the Clinic

Co-chairpersons: Jo Viney (Amgen)Stephan Targan (Cedars Sinai/UCLA)

In this session the audience will begin by learning aboutthe newest family of costimulatory immune regulatorsrecently demonstrated to play a role in intestinal homeo-stasis, as well as the emerging knowledge surrounding thefunction of innate immune receptors at mucosal surfaces.The speakers will also cover the latest informationregarding the elicitation of effector cells and regulatorycells in the intestine, with particular emphasis on theuniqueness of intestinal antigen presenting cells and themajor role that IL-17 plays in the gut. An overview of the

recent experimental therapeutic approaches that havebeen tried in the clinic will provide context for high-lighting the latest breakthrough and potentially promisingtherapies for treating both Crohn*s disease and ulcerativecolitis. Finally, an update on the latest ground-breakinggenetic studies in IBD will provide the foundation forhow genetic information can be transformed into under-standing more about the complex mechanistic basis ofdisease when integrated pathways are studied together,with an emphasis on how this type of approach can revealnovel therapeutic approaches for intervention in thedisease process.

SA01

INTESTINAL IMMUNOMODULATION BY THEEMERGING B7-LIKE BUTYROPHILIN FAMILY

R.M. Swanson, H.A. Arnett and J.L. Viney*

Inflammation Research, Amgen,Seattle, WA and Thousand Oaks, CA

Activation of T cells is known to be modulated bypositive or negative co-signaling molecules. The B7-family of costimulatory molecules has received thegreatest attention so far, and intervention in B7-familysignaling pathways has proven to be an efficaciousstrategy for treating autoimmune diseases in the clinic.Recently, a new family of molecules has garnered interest– the butyrophilins – and early data is suggesting thatthese butyrophilin and butyrophilin-like molecules havethe potential for influencing the nature of immune andinflammatory responses. In vitro studies have revealedthat butyrophilins, such as butyrophilin-like 2 (BTNL2),can negatively regulate T cell proliferation and cytokineproduction. Alterations in intestinal BTNL2 expressionhave been reported in mouse models of IBD as well. And,genetic studies have described polymorphisms in BTNL2which are reported to be associated with disorders such assarcoidosis, myositis and ulcerative colitis. The potentialfor interdicting in the butyrophilin pathways highlightspossible opportunities for developing new therapeuticstrategies for treating autoimmune and inflammatorydisorders. This presentation will focus on the emerginginformation of this new class of regulatory molecules.

SA02

THE ROLE OF ANTIGEN PRESENTING CELLSUBSETS IN REGULATING THE BALANCEBETWEEN IMMUNITY AND TOLERANCE INTHE INTESTINE

Bali Pulendran*, Rosa Maria Salazar, Rajesh Nair,Timothy L. Denning

Emory Vaccine Center, Emory University,954 Gatewood Road, Atlanta, GA 30329, USA

The intestinal immune system must elicit robust immunityagainst pathogens, while restraining reactivity tocommensals and dietary antigens. The mechanisms thatmediate this dichotomy are poorly understood. Ourrecent work has demonstrated a population ofCD11b+F4/80+CD11c- macrophages in the intestinallamina propria, which express several anti-inflammatorymolecules including IL-10, but little or no pro-inflamma-tory cytokines, even upon stimulation with TLR ligands.Furthermore, intestinal macrophages induce FoxP3+

regulatory T cells, via a mechanism dependent on IL-10,and retinoic acid and exogenous TGF-b. In contrast,adjacent CD11b+ dendritic cells stimulate Th17responses, which are suppressed by intestinal macro-phages. Thus, lamina propria macrophages and DCsdifferentially induce T regulatory and Th17 cells, andthe dynamic interplay between these subsets, likely playsa critical role in balancing immune responsiveness versustolerance. Our subsequent analysis is beginning to reveala rich diversity of macrophage and dendritic cell subsets,which seem to be functionally specialized to perform

Page 16: IRA_15th_Conference_21.-24.9.2008

distinct function. Importantly, the representation of thevarious subsets of cells appear to be strikingly different inthe large versus small intestine, and the endogenousmicroflora appear to play a major role in influencing thisrepresentation and function of the antigen presentingcells.

SA03

THE TH17 LINEAGE: DEVELOPMENT,FUNCTION AND REGULATION

Casey T. Weaver*

Department of Pathology, University of Alabama,BBRB 870, 845 19th Street South, Birmingham, AL35294-2170

Na:ve T cells differentiate into effector T cells withenhanced functional potential for orchestrating pathogenclearance largely under the guidance of cytokinesproduced by cells of the innate immune system thathave been activated by recognition of those pathogens.This “secondary” education of post-thymic T cellsprovides a mechanism for appropriately matching adap-tive immunity to cues of the innate immune system.Recently, factors that specify the differentiation of a neweffector T cell subset – Th17 – have now been identified,providing a new arm of adaptive immunity and presentinga unifying model that can explain many heretoforeconfusing aspects of immune regulation, immune patho-genesis and host defense. An update on Th17 develop-ment and function, and consideration of the close links ofTh17 development to regulatory cell development of thispathway will be presented.

SA04

EXPERIMENTAL BREAKTHROUGHTHERAPIES IN IBD

Maria T. Abreu*

University of Miami, Miller School of MedicineMiami, Florida 33101

Understanding the pathogenesis of inflammatory boweldisease (IBD) has formed the basis of therapy for thesecomplex disorders. Immunologically IBD is characterizedby a defective innate immune response and an inappro-priate adaptive immune response to luminal bacterialantigens. Data also suggest a component of defectivebarrier function. Therapies such as trophic growth factorsdirected at barrier function have thus far not beeneffective. At present, monoclonal antibodies are used totarget cytokines important in perpetuation of chronicinflammation. Anti-TNF Abs are effective in the treat-ment of both ulcerative colitis and Crohn*s disease. Twostudies have shown efficacy of an anti-p40 Ab thatantagonizes both IL-12 and IL-23. Given the emergingrole of IL-17 in IBD pathogenesis, this approach is likelyto be effective. Lymphocytes are recruited to the intestinethrough the action of chemokines. Small molecule

antagonists of CCR9, the receptor for CCL25 (TECK),have some benefit in CD. Antibodies against alpha-4integrin are effective in UC and CD. Concern with all ofthese approaches relates to the systemic effects oftherapy, in particular, infectious complications. Finally,mesenchymal stem cells are in clinical trials for CD andmay offer an alternative approach. The biggest advanceswill come when we can personalize therapy based onimmunologic/genetic profiling.

SA05

COMBINATORIAL GENOMICS LEADS TORAPID DISCOVERY OF INTEGRATEDPATHWAYS FOR TARGETED THERAPIES INIBD

Stephan Targan*

Cedars-Sinai Division of Gastroenterology, InflammatoryBowel Disease Center, and Immunobiology Institute,UCLA School of Medicine, Los Angeles CA

In recent years, the scientific community has benefitedfrom an explosion of technological advances in availablemethods to study the genetic variations that exist incommon diseases. For the last two years there have beenmultiple genome wide association studies (GWAS)performed in many common diseases. These have beenable to pinpoint specific biologic processes in specificgenes and genetic variants that may well play a role inthese diseases. A need remains for researchers to conductthese studies on extremely large collections of patients ina given disease entity. Studies of Crohn*s disease, one ofthe two major disease entities which comprise inflamma-tory bowel disease (IBD) have led the way in this newresearch approach. Very recently, a meta analysis ofseveral genome wide association studies have yielded 31confirmed genes associated with Crohn*s disease. It is wellestablished, as evidenced by the many genetically manip-ulated animal models of intestinal inflammation, thatmultiple types of inflammation can be generated by thismeans and suggests functional variants in many differentgenes. Most of these animals generate a dysregulation atthe mucosal surface leading to spontaneous and/orenhanced mucosal inflammation upon perturbation ofinflammation. It is clear that the abnormal interactionsbetween commensal bacterial products and antigens andthe genetically susceptible host is what leads to differentforms of IBD. The major question arising from thesefindings is which of these genetics alterations in animalsare actually relevant to human. An approach for deter-mining human correlates for results generated by animalbased research is to separate the disease process intophysiologic subtypes. We know that the mucosal dysre-gulation in human Crohn*s disease can be defined byspecific immune serologic responses to particular bacte-rial antigens and that these responses can be used to subclassify this disease into more homogeneous groupsranging from very benign to very severe disease thatleads to complications. Very recently, it was determinedthat these serologic responses individually and in combi-nation appear to be representative of mucosal geneticvariants leading to different mechanisms of mucosal

S78 Inflamm. Res., Supplement 2 (2008)

Page 17: IRA_15th_Conference_21.-24.9.2008

inflammatory dysregulation. This advance not only allowstargeted discovery of pathways involved within a defin-able patient population but allows further analysis into

pathogenic interactions, thus, leading to coordinateddiscovery of relevant targets in a given population andfor potentially accelerating novel drug development.

Inflamm. Res., Supplement 2 (2008) S79

Page 18: IRA_15th_Conference_21.-24.9.2008

Tolerance Induction as a Therapeutic Option forAutoimmunity

Co-chairpersons: Steve Nadler (Bristol-Myers Squibb)John Iacomini (Harvard)

The induction of tolerance to autoantigens is the “holygrail” for the treatment of autoimmune disease. Manytherapeutic modalities for tolerance induction have beenstudied over the years, both preclinically and in the clinic.This session will address the basic science and some of themore recent approaches towards the induction of toler-ance including antigen specific tolerization strategies,effector T-cell targeted therapies and utilization of Tregulatory cells.

SA06

WHAT DO T REGULATORY CELLS (TREG) SEEAND DO?

Ethan M. Shevach*

LI/NIAID/NIH, Blg.10, RM11N315, Bethesda, MD,20892, USA

Treg can be identified by the expression of the tran-scription factor, Foxp3, and can be generated both in thethymus and in peripheral lymphoid tissues. TGFb inconcert with IL-2 is a potent induction factor for thedifferentiation of Foxp3+ Treg from na:ve precursors.Polyclonal TGFb-induced Tregs (iTreg) are capable ofpreventing the autoimmune syndrome that develops inscurfy mice that lack Foxp3+ Treg. Antigen-specific iTregcan be used to both prevent and treat autoimmunegastritis that is induced by transfer of na:ve or primedautoantigen-specific T cells. The antigen-specific iTreginhibit the expansion of the effector T cells by disablingthe antigen presenting function of dendritic cells in thelymph node draining the target organ. TGFb complexedwith latency-associated peptide is expressed on thesurface of activated thymus-derived Treg. Coculture ofactivated Treg with na:ve responder T cells results in thede novo generation of fully functional Foxp3+ T cells in acontact-dependent, and TGFb-dependent manner.Generation of functional Foxp3+ T cells via this pathwaymay represent a mechanism by which Treg maintaintolerance and expand their repertoire.

SA07

RESHAPING THE IMMUNE SYSTEM TOINDUCE TOLERANCE IN AUTOIMMUNITYUSING GENE THERAPY APPROACHES

John Iacomini*

Transplantation Research Center, Brigham and Women<sHospital & Children<s Hospital Boston- Harvard MedicalSchool, Boston [email protected].

The principal determining genetic factor in autoimmunetype I diabetes (T1D) is the inheritance of MHC class IIgenes associated with susceptibility to autoimmunedisease. We hypothesized that it may be possible tospecifically address the problem of inheritance of at-riskMHC alleles in T1D by providing genetically predisposedindividuals with MHC genes associated with protectionfrom disease using a gene therapy based approach. Wehave shown that reconstitution of diabetes prone non-obese diabetic (NOD) mice with autologous bonemarrow transduced with retroviruses carrying genesencoding MHC class II molecules associated with protec-tion from T1D, resulting in a state of molecularchimerism, provided robust protection from the develop-ment of diabetes. Induction of molecular chimerism wasalso able to prevent the recurrence of T1D following islettransplantation in NOD mice with pre-existing diabetes.These data suggest that gene therapy based approachescan be used to overcome the underlying problem ofautoimmunity in T1D. These results as well as thedevelopment of additional gene therapy basedapproaches to prevent or cure T1D will be discussed.

Page 19: IRA_15th_Conference_21.-24.9.2008

SA08

AN ANTIGEN-SPECIFIC TOLERANCEAPPROACH FOR THERAPY OF MOUSEMODELS OF MS AND TYPE 1 DIABETES

S. D. Miller*, D. M. Turley, C. Smith, A. Martin, E. Feeney,S. Prasad, D. McCarthy, A. Kohm and X. Luo

Dept. of Microbiology-Immunology, NorthwesternUniversity Medical School, Chicago, IL 60611

Chronic progression of relapsing experimental autoim-mune encephalomyelitis (R-EAE) in the SJL mouse (amodel of MS), and spontaneous diabetes in the NODmouse (a model of type 1 diabetes) are dependent on theactivation of T cells to endogenous tissue epitopes, i.e.epitope spreading. Provided the correct autoepitope(s)are targeted, disease progression in both models can beboth prevented and treated by induction of antigen-specific tolerance using the intravenous injection ofsyngeneic autoepitope-pulsed splenic antigen presentingcells (Ag-SP) fixed with the chemical cross-linker, ethyl-ene carbodiimide (ECDI). Our results indicate that theinsulin B chain epitope (Ins B9-23) is the initiatingdiabetogenic epitope in NOD mice as tolerance inducedwith either intact insulin (Ins-SP) or Ins B9-23 (InsB9-23-SP), but not with GAD65509-528, GAD65524-543, or IGRP206-

214 coupled splenocytes at 4-6 weeks of age inhibitsdiabetes development. In contrast, regulation of newonset diabetes in 20 week old NOD mice is only inducedby tolerization with Ins-SP, not InsB9-23-SP, suggestingspreading to a distinct insulin epitope. Ag-SP tolerance isdue primarily to re-presentation of apoptotic Ag-SP byhost splenic plasmacytoid dendritic cells which induceboth PD-1-dependent clonal anergy and the activationantigen-specific Foxp3+ regulatory T cells (Tregs).

SA09

TEPLIZUMAB, A HUMANIZED, FC RECEPTOR(FCR) NON-BINDING ANTI-CD3 MONOCLONALANTIBODY IN RECENT-ONSET TYPE 1DIABETES (T1DM)

Ronald L. Wilder*

MacroGenics, Inc., 1500 East Gude Drive, Rockville, MD,20850

Teplizumab, which is also called hOKT3g1 (Ala-Ala) andMGA031, have the same binding specificity and avidityfor CD3 epsilon as murine OKTG3, but the Fc componentof the antibody does not bind FcR or activate comple-ment. Humanization and the changes in the Fc compo-nent of teplizumab have resulted in profoundly differentbiological activity compared to the parent monoclonalantibody. It is 1000-10,000-fold less mitogenic, is onlyweakly activating and does not appear to engage costi-mulatory signals. Data available to date suggest thatteplizumab inactivates/anergizes T effector cells andexpands T regulatory cells. Its capacity to induce acytokine-release syndrome is dramatically bluntedcompared to OKTG3. In addition, teplizumab does notproduce long-term lymphopenia. It has potential appli-cations in a wide spectrum of autoimmune diseases andtransplant indications. Published data have indicated thatteplizumab has the capacity to preserve and possiblyimprove pancreatic islet cell function in subjects withrecent-onset T1DM. It is under evaluation in 3 trials forthis indication, i.e. , two phase 2 trials (the Abate andDelay trials), and a phase 2/3 trial (the ProtHgH trial).These trials are actively enrolling.

Inflamm. Res., Supplement 2 (2008) S81

Page 20: IRA_15th_Conference_21.-24.9.2008

Inflammation and Pain

Co-chairpersons: Jane Connor (Medimmune)Jeff Mogil (McGill U.)

Pain (along with redness, heat and swelling) is one of thefour hallmarks of inflammation. However, in spite ofwell-established animal models of inflammatory pain(such as carrageenan-induced hyperalgesia) providingconfidence in rationale and currently marketed clinicaltherapies (such as NSAIDs and COX-2 inhibitors) in thisarea, there continues to be a significant unmet medicalneed for the treatment of pain driven by inflammation.This symposium will focus on more recent approaches toevaluating pain in animals as well as novel targetsrecently identified that possess the potential to treatthose patients whose pain goes untreated in spite ofcurrent therapies.

SA10

THE SURPRISING COMPLEXITY OF PAINTESTING IN THE LABORATORY MOUSE

Jeffrey S. Mogil*

Dept. of Psychology, McGill University, 1205 Dr. PenfieldAve., Montreal, QC H3A 1B1 CANADA

Researchers studying pain in animal models face aconundrum. Any number of assays are available withhigh face validity, but the mechanisms underlying painprocessing in these assays are more and more understoodto be of little relation to mechanisms underlying clinicalpain. However, the production of more clinically relevantpain states in laboratory animals yields few scorabledependent measures. As a result, most pain researchersrely on the measurement of reflexive, evoked hyper-sensitivity responses after neuropathic or inflammatoryinjury, whereas clinical pain in humans features muchspontaneous pain and an important cognitive andemotional overlay. Making matters worse, it has becomevery clear that pain can be modulated both quantitativelyand qualitatively by factors such as genetic background,sex, arousal, social factors and laboratory environment.Such complexities have served to discourage measure-ment of pain in inflammatory models; this is unfortunatebecause the rather poor correlation between inflamma-tion and pain in these states suggests that pain really doesneed to be studied separately. The challenges and promise

of pain testing in the laboratory mouse will be discussed,with a focus on inflammatory pain.

SA11

ROLE OF ASICS IN MUSCLE AND JOINTHYPERALGESIA ASSOCIATED WITHINFLAMMATION

K.A. Sluka*, R.Y. Walder, L.A. Burnes, S.J. Kolker, andM. Ikeuchi

Physical Therapy and Rehabilitation Science,University of Iowa, Iowa City, IA 52242

Acid Sensing Ion Channels (ASICs) are important forsensing decreases in pH and are found on nociceptiveafferents. Using ASIC1 and ASIC3 knockout mice, wemechanical sensitivity of the muscle or joint (primaryhyperalgesia) and of the paw (secondary hyperalgesia)before and after induction of inflammation with carra-geenan. In ASIC3 knockout mice, withdrawal thresholdsof the muscle and joint decreases similarly to wild-typemice after induction of inflammation. However in ASIC1knockout mice, withdrawal threshold of the muscle didnot decrease after induction of inflammation. In contrast,ASIC3 knockout mice did not show decreases in with-drawal thresholds of the paw; ASIC1 knockout mice stillshowed increased mechanical sensitivity similar to wild-type controls. A non-selective ASIC antagonist, A-317567, reverses both the primary and secondary hyper-algesia in wild-type mice. In ASIC3 knockout mice theprimary hyperalgesia is also reversed by A-317567 and inASIC1 knockout mice the secondary hyperalgesia isreversed by A-317567. Further there is an upregulation ofASIC3 in primary afferent fibers innervating joint, and inDRG retrogradely labeled from joint. This upregulationin DRG occurs in non-peptidergic neurons. Thus ASICsare upregulated by inflammation; ASIC3 appears to beimportant for development of secondary hyperalgesia;and ASIC1 appears to be important for development ofprimary hyperalgesia. Supported by National Institutes ofHealth AR053509.

Page 21: IRA_15th_Conference_21.-24.9.2008

SA12

PRONOCICEPTIVE ACTIONS OF CCL2 ANDCXCL12 AFTER PERIPHERAL NERVE INJURY

Fletcher A. White*

Department of Cell Biology, Neurobiology & Anatomy,Anesthesiology Research Laboratory, Burn and ShockTrauma Institute, Loyola University-Chicago,Maywood, IL 60153

C-C and C-X-C chemokines and their receptors may playcrucial roles in the pathophysiology of pain. Evidence of apronociceptive contribution includes a delayed upregula-tion of monocyte chemoattractant protein-1 (MCP-1/CCL2) and its respective receptor, CCR2, in both injuredand adjacent, non-injured sensory neurons. Activation ofneuronal CCR2 by CCL2 elicits membrane depolariza-tion, triggers action potential and sensitizes nociceptorsvia transactivation of transient receptor potential chan-nels, TRPA1 and TRPV1. Peripheral nerve injury alsoproduces a latent increase in both neuronal expression ofthe chemokine, stromal-derived factor-1 (SDF1/CXCL12)and functional signaling by its cognate receptor, CXCR4.Treatment of nerve injured rodents with a CCR2 receptorantagonist, ((R)-4-Acetyl-1-(4-chloro-2-fluorophenyl)-5-cyclohexyl-3-hydroxy-1,5-dihydro-2H-pyrrol-2-one, atpost-injury day (PID) 14 or 28, but not PID 7, temporarilyattenuates hypernociceptive pain behavior. The non-peptide CXCR4 receptor antagonist, AMD3100,produces similar reversal of hypernociception in therodent at PID 28, but not PID 14. Taken together,chemokine receptor antagonists may be important ther-apeutic interventions for pain syndromes. Funded by NIHRO1NS049136, R01NS043095; Falk Medical ResearchTrust.

SA13

BOTH EP4 ANTAGONISTS AND MPGES-1INHIBITORS RELIEVE PAIN IN RODENTMODELS OF ARTHRITIS

Daigen Xu*, Bernard CDtE, Yongxin Han, YvesDucharme, Richard W. Friesen, Joseph Mancini, DenisRiendeau, Laurent Audoly, and Alex Therien.

Merck Frosst Centre for Therapeutic Research,16711 Transcanada Hwy,Kirkland, Qc, Canada, H9H 3L1

Traditional non-steroidal anti-inflammatory drugs(NSAIDs) and cyclooxygenase-2 (COX-2) inhibitorsprovide pain relief in arthritis by blocking the synthesisof prostaglandins (PGs). Recent evidence demonstratesthat deletion of the EP4 receptor or microsomal PGEsynthase 1 (mPGES-1) renders mice resistant to auto-immune arthritis, similarly to the deletion of COX-2,suggesting that PGE2 is a major pro-inflammatoryprostaglandin in arthritis and mPGES-1 or EP4 may bea useful target for the treatment of the disease. In supportof the hypothesis, we demonstrated that a selective EP4antagonist was as effective as a COX-2 inhibitor or anNSAID at mitigating adjuvant-induced arthritis in ratsand relieving iodoacetate-induced osteoarthritic pain inguinea pigs. We also noted a similar analgesic efficacy bya selective mPGES-1 inhibitor in guinea pigs. UnlikeNSAIDs, both the EP4 antagonist and the mPGES-1inhibitor were well tolerated by the gastrointestinal tract,causing no mucosal erosions or leakage. Together, thesefindings suggest that pharmacological inhibition of PGE2

synthesis or activity is a useful approach for treatinginflammatory diseases.

Inflamm. Res., Supplement 2 (2008) S83

Page 22: IRA_15th_Conference_21.-24.9.2008

Biologics as a Platform for Inflammation Therapies

Co-chairpersons: John Beals (Lilly)Frank Castellino (Notre Dame)

Biological therapeutics are increasingly being used totreat diseases that are exacerbated by inflammation, e.g.,rheumatoid arthritis (anti-TNFa and IL-1 antagonisttherapies) and sepsis (activated Protein C). The goal ofthis session is to explore the hurdles associated with usingbiologicals as inflammation therapies and solutions toaddress these challenges.

SA14

THE PROTEIN C SYSTEM RESIDES AT THECROSSROADS OF HEMOSTASIS ANDINFLAMMATION

Frank Castellino*

WM Keck Center for Transgene Research, 230Raclin-Carmichael Hall, University of Notre Dame,Notre Dame, Indiana 46556

Abstract not available at time of printing, see Supple-ment.

SA15

BIOLOGICAL THERAPEUTICS, T EFFECTORAND T REGULATORY CELLS IN AUTOIMMUNEDISEASE

Anne S. De Groot*

CEO/CSO, EpiVax, Inc., Providence, RI 02903; Institutefor Immunology and Informatics, Brown UniversityMedical School Providence, RI 02912, USA

Immunogenicity elicited by protein therapeutics cancause serious side effects in humans. Tools designed byexpert immunoinformaticians have enabled the predic-tion of immunogenicity and the prospective identificationof subjects who may be at increased risk of developingadverse immune responses. These same techniques canalso be used to elucidate the dynamic balance between Teffector and T regulatory cells in the development andtreatment of autoimmune diseases. We have used HLAtyping in conjunction with EpiMatrix, an in-silico epitope-

mapping tool, to predetermine the immunogenicity ofbiological therapeutics. In a recent published case, weidentified promiscuous epitopes (“EpiBars”) in the C-terminal region of a recombinant fusion protein(FPX)(1). On administration of FPX in 76 healthyhuman subjects, 37% developed antibodies after a singleinjection; immune responses were limited to individualswho had the HLA that could present the immunogenicregions of the peptide. A memory T-cell response againstthe carboxy-terminus of the peptide was both predictedand observed, and also as predicted, HLA-haplotypeDRB1*0701/1501 was associated with the highest T-celland antibody response. For monoclonal antibodies, aclose correlation between silico immunogenicity assess-ment and the confirmed immunogenicity of monoclonalantibodies in the clinic has also been documented(2). Insilico tools such as Optimatrix can be used in combinationwith in vitro/in vivo approaches to diminish the T cellepitope content of monoclonal antibodies and proteintherapeutics. In addition, exploitation of T regulatory cellsuppression of immune response may be another meansof suppressing T dependent antibodies to protein ther-apeutics. The latter approach, which has been the mainfocus of our work for the last 18 months, also holds somepromise for the treatment of autoimmune diseases. Thispresentation will illustrate the use of these readilyavailable tools to pre-determine immunogenicity. Techni-ques for salvaging immunogenic therapeutics will also beaddressed.

1. Koren E, De Groot AS, et al. Clinical validation of the”in silico” prediction of immunogenicity of a humanrecombinant therapeutic protein. Clin Immunol. 2007May 7; epub. doi:10.1016/j.clim.2007.03.544.

2. Hai S, McMurry JA, Knopf P, Martin W, De Groot AS.Immunogenicity screening using in silico methods:Correlation between T-cell epitope content and clin-ical immunogenicity of monoclonal antibodies. InTherapeutic Antibodies: from Theory to Practice.Zhiqiang An, Editor. John Wiley and Sons. (Scheduledfor publication Fall 2008).

Page 23: IRA_15th_Conference_21.-24.9.2008

SA16

CHARACTERIZATION AND OPTIMIZATION OFMONOCLONAL ANTIBODIES FOR THETREATMENT OF INFLAMMATORY DISEASES -CASE STUDIES

Jirong Lu*

Lilly Research Laboratories, A Division of Eli Lilly &Company, Lilly Corporate Center, Indianapolis, IN 46285

LY634923 is an interleukin-1b (IL-1b) antagonist anti-body Deamidation of Asn55 (-PGNGNI-) in the CDR2of the heavy chain was identified. This site is readilydeamidated at neutral pH and presents challenges for thedevelopment of this antibody as a therapeutic agent. Tobetter understand the impact of deamidation on antigenbinding, site-directed mutagenesis was used to replaceAsn55 and Gly56 to eliminate or slow-down deamidation.Ten analogs, together with the wild-type molecule, wereproduced and evaluated. Substitution of Asn55 elimi-nated chemical instability but resulted in a 3 to 200-folddecrease in binding affinity to IL-1b that translated intolower neutralization activity in vitro. Both charge and sizeof the side chain at position 55 appeared to impactbinding of the antibody to IL-1b. The analog Asn55Asp,which mimics fully deamidated form, displayed a 20-foldreduction in activity. Mutation of Asn55 to Ser, Ala orThr has the lowest impact on binding (~3-fold). Incontrast to limited site-directed mutagenesis, whendirected evolution in vitro was used to evaluate theimpact of all possible amino acid substitutions at everyposition throughout the CDR, a number of antibodyvariants with improved stability and enhanced affinitywere generated.

SA17

BIOLOGICAL THERAPEUTIC APPROACH TOATHEROSCLEROSIS: CHALLENGES ANDOPPORTUNITIES, TWO CASE STUDIES INEXPERIMENTAL DISEASE

Godfrey S. Getz*

Departments of Pathology, Biochemistry, and MolecularBiology, The University of Chicago, 5841 S. MarylandAvenue, MC 1089, Chicago, IL 60637-1470

Atherosclerosis is a chronic inflammatory state of varioussites in large and medium size arteries. It is the pathology

that underlies much of the cardiovascular disease andmorbidity found in a major portion of the first worldpopulations. I will briefly review the pathogenesis ofatherosclerosis as studied in mouse models of the disease.It is clear that it results from increases in plasma lipidsand other risk factors and from local inflammatoryreactions in the vessel wall itself. Much of the currentpreventative approach relies on agents that lower plasmacholesterol and LDL in particular coupled with attemptsto increase HDL. Our research is directed at two majorquestions: how does the adaptive immune system influ-ence atherosclerosis and how does HDL compositioninfluence the anti-inflammatory role of this lipoprotein.In studying the immune system modulation of athero-sclerosis we have noted in a number of cases that variousvascular sites are differentially influenced by immuneregulators. This poses a significant challenge in usingimmune modulation as a window to an approach to theprevention and treatment of the vascular disease.

We have recently found a close connection between theimmune system and lipoprotein homeostasis. This ismediated by the co-stimulatory set of molecules (secondsignals in antigen stimulation) of the TNF superfamilyand LIGHT and lymphotoxin in particular, expressed onthe surface of T cells. These two membranes boundcytokines signal via the lymphotoxin beta receptorexpressed on among other cells, the hepatocytes. Inter-diction of this pathway with soluble synthetic receptorlowers plasma lipids. This is mediated by effects on theenzyme, hepatic lipase which may function either as anenzyme or as a clearance receptor ligand. We are studyingthe particular role of NKT cells in this regulatorynetwork. The latter cells are enriched among the lympho-cytes of the liver. This pathway and the potential of thesoluble receptor as a therapeutic will be reviewed.

HDL is thought to be therapeutic. The major protein ofHDL is apolipoprotein A-1 and overexpression of thelatter results in atheroprotection. There has been greatinterest recently in the use of synthetic apolipoprotein A-1 mimetics to treat atherosclerosis. These agents havemany potential actions in mediating cholesterol removalfrom the vessel wall macrophages and in their potential asantioxidants and anti-inflammatories.

I will review our studies on several variants of themimetics as anti-oxidative, anti-inflammatory and anti-atherogenic agents in animals with the usual HDL andthose lacking HDL. (This work is supported by grantsfrom the National Heart, Lung and Blood Institutes ofthe NIH).

Inflamm. Res., Supplement 2 (2008) S85

Page 24: IRA_15th_Conference_21.-24.9.2008

The IL-17/IL-23 Axis in Autoimmunity

Co-chairpersons: Joel Tocker (Amgen)Sarah Gaffen (U. Pittsburgh)

The regulation of IL-17 production from a distinct andpathogenic population of T helper cells and the contri-bution of these cells and IL-17 in animal models ofautoimmune disease and inflammation is a hot topic.Further the complexities in the regulation of IL-17production, the IL-17R signaling complex, and the natureof the IL-17 ligand itself offer numerous approaches andopportunities to modulate IL-17 biologic activity.

SA18

DISSECTING THE IL-17 RECEPTOR: SIGNALINGAND FUNCTION

Sarah L. Gaffen*

Dept of Oral Biology, SUNY Buffalo, NY; Division ofRheumatology & Clinical Immunology, University ofPittsburgh, PA

IL-17 is the hallmark cytokine of a newly-discoveredinflammatory T cell subset termed “Th17.” IL-17 hasbeen shown to mediate disease pathology in a variety ofautoimmune conditions including rheumatoid arthritis(RA). Not surprisingly, blockade of IL-17 or its receptoris effective in limiting disease in several animal models ofRA, and trials in humans are now ongoing. The IL-17receptor is the founding member of a new superfamily ofcytokine receptors that bears little homology to otherfamilies of cytokine receptors. Therefore, few predictionsregarding signaling mechanisms can be made simply onprimary sequence. Our laboratory has established systemsto define structure-function relationships in the IL-17receptor complex in terms of receptor subunit assemblyand downstream signal transduction. Here, I will discussrecent progress in defining the important structural andfunctional elements that dictate how this unique receptorsystem operates.

SA19

THE IDENTIFICATION OF A HUMANIL-17F/IL-17A HETERODIMER AND THERECEPTOR COMPLEX UTILIZED INSIGNALING

Jill Wright*

Inflammation Department, Wyeth200 Cambridge Park Drive, Cambridge, MA 02140

IL-17A and IL-17F are members of the IL-17 pro-inflammatory cytokine family and have been linked to avariety of inflammatory and autoimmune conditions. IL-17A and IL-17F signal through a receptor complex thatconsists of IL-17RA and IL-17RC. The crystal structureof IL-17F shows that IL-17F forms a disulfide-linkeddimer that contains a cysteine knot motif. Given the highdegree of amino acid homology between IL-17A and IL-17F and the conservation of the four cysteines that formthe knot, it is likely that IL-17A and IL-17F adopt asimilar structure. We hypothesized that IL-17F and IL-17A could form a heterodimer due to their sequencehomology and overlapping pattern of expression. In thissymposium, I will present data that shows that activatedhuman CD4+ T cells not only produce the IL-17A andIL-17F homodimers but also produce the IL-17F/IL-17Aheterodimer that utilizes the same receptor complex asIL-17A and IL-17F homodimers.

SA20

THE IL-23/IL-17 AXIS IN ARTHRITIS

Erik Lubberts*

Erasmus Medical Center, Rotterdam, The Netherlands([email protected])

Interleukin-17A (IL-17A) contributes to the pathogenesisof arthritis. Data from experimental arthritis indicate IL-17 receptor signaling as a critical pathway in turning anacute synovitis into a chronic destructive arthritis. Theidentification of six IL-17 family members (IL-17A-F)may extend the role of this novel cytokine family in thepathogenesis of chronic destructive joint inflammation.Whether the successful anti-IL-17A cytokine therapy inmurine arthritis can be effectively translated to human

Page 25: IRA_15th_Conference_21.-24.9.2008

arthritis need to be tested in clinical trials in humans.Interestingly, IL-17A and IL-17F are secreted by thenovel T helper subset named Th17. This novel pathogenicT cell population induces autoimmune inflammation inmice and is far more efficient at inducing Th1-mediatedautoimmune inflammation in mice than classical Th1 cells(IFN-g). IL-23 plays a critical role in Th17 survival andactivity in experimental autoimmune models. Studies willbe discussed to show the role of IL-23 in IL-17/IFN-gamma subgroup polarization in the autoimmunecollagen-induced arthritis and its role in chronic non-autoimmune arthritis. In addition, modulation of Th17polarization in experimental mouse models as well as inearly rheumatoid arthritis patients will be presented.

SA21

STAT TRANSCRIPTION FACTOR SIGNALING INMOUSE AND HUMAN TH17 POLARIZATION

Arian Laurence*

NIH, NIAMS, Lymphocyte Cell Biology Section10 Center Drive, Rm 13C120-9B, Bethesda, MD, 20892USA

Studies in mice have shown that T helper (Th) celllineages are critical in defending against classes ofpathogens: Interferon gamma (IFNg) producing Th1cells for defense against viruses and interleukin-4 (IL-4)producing Th2 cells for defense against nematode worms.The novel IL-17 producing Th17 cells are critical fordefense against bacterial infections, and mice that lack theability to make IL-17 have difficulty in forming abscessesand overcoming bacterial pneumonia. Studies in Stat3knockout mice have shown that this transcription factor isnecessary for the formation of Th17 cells. Several groupshave documented the key cytokines required for theinduction of Th17 cells in both mice and humans yet thereremains disagreement as to the correct cocktail in thelatter. Despite the controversy, a common feature of allthe human studies reported is that a Stat3 activatingcytokine is required. Patients with Job syndrome, aninability to defend against bacterial infections, havemutations in their Stat3 gene and are a unique oppor-tunity to study the role of Stat3 in human Th cellpolarization. Job syndrome patients are unable to makeTh17 cells. Further studies are required to determine howthis contributes to the patho-physiology of the syndrome.

Inflamm. Res., Supplement 2 (2008) S87

Page 26: IRA_15th_Conference_21.-24.9.2008

Novel Therapeutics

Co-chairpersons: Bruce Tomczuk (Chemnomics LLC)Rey Panettieri (U. Pennsylvania)

This symposium will present novel therapies for Asthma,COPD, Crohn*s Disease/Psoriasis, and RheumatoidArthritis. The Asthma presentations will focus on apharmacological (IL-4 receptor antagonist, AMG-317, inPhase II) and a non-pharmacological (Bronchial Ther-moplasty) approach toward this complex disease. Due tothe high unmet medical need, the presentation on COPDwill provide an early stage opportunity for the sympto-matic relief of mucus production. A late-stage clinicalcandidate, ABT-874, for Crohn*s Disease (Phase II) andPsoriasis (Phase III) targets the inflammatory cytokines,IL-12 and IL-23 at their shared IL-12 receptor beta 1. Atherapeutic for rheumatoid arthritis (Baminercept inPhase IIb) targets the lymphotoxin-b/LIGHT pathwayfor inhibition of lymphoid structures in inflamed joints.An additional talk will cover late-breaking clinical results.

SA22

INHIBITION OF IL-4Ra : A PROMISINGTHERAPEUTIC STRATEGY FOR ASTHMA

Joel Tocker1, Michelle Horner2, Michael Vincent2,Christopher Banfield2, Shao-Lee Lin2*

1Amgen Inc., Seattle, WA, 2Amgen Inc., One AmgenCenter Drive, Rm 38-2A-4-1, Thousand Oaks, CA 91360

The cytokine interleukin-4 (IL-4) plays a key role in thedevelopment of type 2 helper T lymphocyte (Th2)immune responses. Th2 mediated immunity underliesallergic inflammation and protective immunity to extrac-ellular organisms. Other cytokines associated with Th2immune response include IL-13, which also signalsthrough the IL-4 receptor alpha (IL-4Ra) complex.Evidence from murine models implicates IL-4 and IL-13in the induction and maintenance of asthma. The efficacyof IL-4R antagonism was demonstrated with a surrogateantibody in mouse models of allergic inflammation. In acockroach allergen re-challenge model in mice, treatmentwith Mu317RaXMu reduced lung inflammation, airwayhyperresponsiveness, total serum IgE, and accumulationof whole lung hydroxyproline. In cynomolgus monkeys,weekly administration of Cy317HuXCy was well toler-ated in 4-, 13-, and 26-week repeated dose safety studies.AMG 317 is a fully human monoclonal antibody being

investigated in asthma based on its ability to inhibit IL-4and IL-13 signaling through IL-4Ra. In Phase 1 studies,no clinically relevant safety signal was evident followingadministration of single doses of AMG 317 up to 1000 mg.Proof of concept for IL-4Ra blockade has been demon-strated in inhaled allergen challenge studies of Pitrakinra,a molecule that blocks IL-4 and IL-13 through IL-4Ra.Results showed a reduction in the decrease in FEV1

during the late phase response, and decreased FeNOpost-treatment. AMG 317 is currently being evaluated ina Phase 2 dose ranging study in subjects with moderate tosevere atopic asthma.

SA23

POTENTIAL FOR BRONCHIALTHERMOPLASTY TO REVERSE AIRWAYREMODELING ASSOCIATED WITH ASTHMA

Gerard Cox*

Firestone Institute for Respiratory Health,McMaster University, East Hamilton, ON, L8N 4A6,Canada

Chronic asthma is associated with remodeling of thebronchial wall and especially with increased mass andactivity of smooth muscle that is found in airways.Medical therapies can address many of the key featuresof asthma, such as bronchoconstriction and airwayinflammation, but none of the currently available thera-pies can reverse remodeling. We have explored a physicalapproach, using direct thermal treatment of the bronchi,to reduce the mass of airway smooth muscle and thusreduce the potential for bronchoconstriction. The AlairG

system comprises a radiofrequency generator thatdelivers energy through a specially designed catheterthat has at the distal end an expandable basket of 4electrodes. When positioned at the time of bronchoscopy,the electrodes are expanded to contact the airway walland a controlled bolus of energy is supplied over 10seconds that achieves a target temperature in the airwaywall. In a series of studies in canine airways, we found a“dose-response” effect of temperature on subsequent lossof airway muscle mass, and on reduced bronchoconstric-tion in response to directly administered methacholine.Furthermore, the reduction in methacholine-inducedbronchoconstriction correlated with the degree to which

Page 27: IRA_15th_Conference_21.-24.9.2008

smooth muscle was diminished. These effects persistedout to three years.

In 3 clinical trials in patients with a range of asthmaseverity from mild to severe/steroid-dependent, we foundthat bronchial thermoplasty led to improvements insubjective and objective measures of asthma controlincluding improvements in peak flow readings andreduced need for symptom-relieving medications. Thesebenefits could be related to reduced activity of airwaysmooth muscle. The benefits of bronchial thermoplastyare most evident in those with the most severe asthma.While bronchial thermoplasty is associated with frequentshort term increase in airway symptom, no long termadverse effects have been found.

SA24

BIO-11006: TARGETING THE MARCKS PROTEININ COPD

Kenneth B. Adler*

Dept. of Molecular Biomedical Sciences,North Carolina State Univ., 4700 Hillsborough St, Raleigh,NC 27606, USA

Work from this laboratory and from others has shownthat MARCKS protein (myristoylated alanine-rich Ckinase substrate) has an integral role in two componentsof chronic bronchitis: inflammation and mucus secretionin the airways. A peptide related to the N-terminus ofMARCKS (the MANS peptide) had dramatic amelio-rating effects in both in vitro and in vivo models of mucinsecretion and inflammation. Here, we report that asoluble analog of MANS, BIO-11006, produced byBioMarck, RTP, North Carolina, has similar effects in avariety of model systems that relate to asthma andchronic bronchitis. BIO-11006 attenuated, in a concen-tration-dependent manner, both mucin secretion andairway obstruction in mice sensitized to OVA and treatedwith MCH.. While all doses ranging from 1-20 mM werefound effective, 10 mM solution aerosolized for 30 minwas found to be most effective. The effect of BIO-11006was maximal immediately after the aerosol was admin-istered and declined thereafter with t1/2 of 2-3 hrs.Statistically and biologically significant effects were seenas late as 4 hrs after pre-treatment. BIO-11006 alsoshowed significant effects in two different mouse modelsof chronic bronchitis: it markedly inhibited ozone-induced neutrophil migration into the airways and alsoinhibited expression of cytokines such as IL-6, TNFa andKC (the murine equivalent of IL-8). In addition, it hadsignificant inhibitory effects on mucin secretion in micetreated with human neutrophil elastase. Collectively, theresults indicate that BIO-11006, a peptide related to

MARCKS protein, possesses potent anti-mucus and anti-inflammatory properties. BIO-11006 currently is in Phase2 clinical trials in COPD patients.

SA25

ANTI-IL-12/IL-23 TREATMENT FOR CROHNASDISEASE AND PSORIASIS

Trudi Veldman*

Abbott Bioresearch Center100 Research DriveWorcester, MA 01605

The anti-IL-12/23p40 antibody ABT-874 is a fully humanrecombinant antibody isolated using phage display tech-nology. Abbott has completed several clinical studies withABT-874 in Crohn*s disease, multiple sclerosis andpsoriasis. The clinical data from the Phase 2a Crohn*sdisease study and the 12-week Phase 2 psoriasis study willbe presented. ABT-874 was significantly more efficaciousthan placebo in the treatment of moderate to severeplaque psoriasis, and appears to have a favorable safetyprofile. A potential mechanism that may explain theextended pharmacodynamic effect of drug treatment willbe discussed.

SA26

BAMINERCEPT: TARGETING LT-b AND LIGHTPATHWAY IN RA

Tony Manning*

Inflammation & Autoimmune Diseases Research,BiogenIdec, 14 Cambridge Center,Cambridge, MA 02142

Lymphotoxins (LT-a and LT-b), LIGHT [homologous toLT, inducible expression, competes with herpes simplexvirus (HSV) glycoprotein D for HSV entry mediator(HVEM), a receptor expressed on T lymphocytes], andtheir specific receptors LTbR and HVEM, are membersof the immediate family of the larger TNF superfamily.These cytokines establish a critical communication systemrequired for the development of secondary lymphoidtissues. Such reactions are hypothesized to play animportant role in autoimmune diseases such as rheuma-toid arthritis and systemic lupus erythematosus. Thispresentation will discuss the pre-clinical and emergingclinical evidence suggesting that targeting LTbR willmodulate disease-related biology and provide clinicalbenefit in rheumatoid arthritis.

Inflamm. Res., Supplement 2 (2008) S89

Page 28: IRA_15th_Conference_21.-24.9.2008
Page 29: IRA_15th_Conference_21.-24.9.2008

Mini-symposia and Poster Session Abstracts

Page 30: IRA_15th_Conference_21.-24.9.2008
Page 31: IRA_15th_Conference_21.-24.9.2008

A100

NOCICEPTIN ANTAGONISTS FOR THETREATMENT OF INFLAMMATORY BOWELDISEASE

Carsten Alt*, Ken Shew, Than Thuy Tran, Willma Polgar,Lawrence Toll, and Annalisa D�Andrea

Clinique Laboratories, Melville, NY

Inflammatory bowel disease (IBD) is represented by acomplex of heterogeneous gastrointestinal diseases thatinclude two major entities, ulcerative colitis and Crohn�sdisease. Nociceptin is a neuropeptide that has beenhypothesized to be a stimulator of inflammation, andthe Nociceptin receptor is expressed at high levels in thegut. In this study we evaluated the prophylactic andtherapeutic potential of Nociceptin antagonists in themouse model of DSS-induced colitis. To this end weinduced colitis by feeding C57BL/6 mice with 3.5% DSSsolution in the drinking water for 5 subsequent days.Nociceptin antagonists were administered intraperito-neally starting at day 0 until sacrifice at day 12. Ourresults show that treatment with Nociceptin antagonistsdramatically ameliorated the development of colitis asobserved by changes in body weight, fecal consistency,and colon length. Additional studies are in progress toevaluate the mechanism of action of nociceptin antago-nists during IBD.

Our results show that treatment of the animals withNociceptin antagonists significantly ameliorates the DSS-induced colitis, suggesting that inhibition of Nociceptinmight be an effective therapy for IBD.

A101

DESIGN, SYNTHESIS AND BIOLOGICALEVALUATION OF AZOLE DERIVATIVES OFDIPHENYL ACETIC ACID ASANTI-INFLAMMATORY AGENTS

Mohd. Amir*, M. K. Saifullah, Israr Ali and M. S. Zaman

Department of Pharmaceutical Chemistry, Faculty ofPharmacy, Hamdard University, New Delhi – 110062,India

Long-term use of arylalkanoic acids such as ibuprofen,diclofenac and flurbiprofen has been associated withgastrointestinal ulceration, bleeding and nephrotoxicity.To reduce their gastric toxicity a number of derivativeshave been prepared and in some of these carboxylicgroup has been replaced by five membered heterocyclicmoieties such as 1,3,4-oxadiazole/thiadiazole and 1,2,4-triazole. In our attempt to discover new and useful agentsfor treating inflammatory diseases, we have replaced thecarboxylic acid group of diphe- nyl acetic acid with theseheterocyclic moieties. The cyclized derivatives werescreened by carrageenan induced rat paw edema methodand showed 49.03% to 83.02% inhibition, where asstandard drug ibuprofen showed 74.71% inhibition atthe same oral dose. Compounds showing significant anti-

inflammatory activity were selected to study their anal-gesic, ulcerogenic and lipid peroxidation activities. Thetested compounds showed reduction in ulcerogenicactivity compared to ibuprofen. The compounds thatshowed less ulcerogenic effect also produced less malon-dialdehyde (MDA) content in gastric mucosa, which isone of the end products of lipid peroxidation.

A102

INFLAMMATORY CHANGES INSCHIZOPHRENIA WITH ANTIPSYCHOTICTREATMENT

R. A. Baillie*, J. McEvoy, R. Kaddurah-Daouk

Rosa & Co, Duke University Medical Center, Durham,NC, USA

Patients with schizophrenia (SCH) are at increased risk ofdeveloping metabolic disease. Furthermore, some atyp-ical antipsychotics are associated with increased risk ofweight gain and metabolic changes, increasing the risk formetabolic disease. Previous research indicated that bothschizophrenia and atypical antipsychotic treatment modu-lated phospholipid concentrations and apparent produc-tion of arachidonic acid, a critical precursor for theinflammatory lipids. We used a targeted metabolomicapproach to evaluate the inflammatory lipid and cytokinechanges occuring in schizophrenia and with antipsychotictreatment. Plasma concentrations of specific prostaglan-dins (PG) and other inflammatory lipids were decreasedin patients with schizophrenia compared to controlsubjects. Plasma concentrations of PGD2 were decreasedafter treatment with antipsychotics. Adipokine concen-trations, including adiponectin and PAI1 were different incontrols vs. schizophrenia subjects and were furtherchanged by antipsychotic treatment. Inflammationmight be an important common process relating schizo-phrenia and antipsychotic treatment to the developmentof weight gain and metabolic disease.

A103

PHARMACOLOGICAL CHARACTERIZATION OFA NEW MODEL OF CYNOMOLGUS SKININFLAMMATION MEASURED WITH NOVELMETHODOLOGIES

A.E. Berson*, M. Brovarney, C.P. Mao, I. BaileyHealy, N.Poy, P. Ravindran, M.E. Fuentes, and K. Dabbagh

Roche Palo Alto, Palo Alto, CA 94303

The development of a novel delayed-type hypersensitivity(DTH) in Cynomolgus monkeys was used to assess therole of T-cells in skin inflammation. The inflammationwas scored by traditional DTH methods, includingimmunohistochemistry. Further analysis of skin biopsiesby gene expression provided a more through and efficientcharacterization of the response that correlated with theDTH response. The skin inflammation was blocked byDexamethasone but not FK506, revealing a surprising

Inflamm. Res., Supplement 2 (2008) S93

Page 32: IRA_15th_Conference_21.-24.9.2008

lack of effect of T cell blockade in the model. This datademonstrates a minimally invasive and rapid method tofully characterize skin inflammation in non-humanprimates that has the potential for translation to humans.

A104

BIACORE ANALYSIS REVEALS DISTINCTRECEPTOR (a1 AND a2) BINDING INHIBITIONCHARACTERISTICS OF ANTI IL-13 ANTIBODIES

Sahana Bose*, Chengbin Wu, Zehra Kaymakcalan andDenise Karaoglu Hanzatian

Abbott Bioresearch Center, 100Research Drive, Worcester,MA, 01605

Human IL13 is a glycoprotein, which plays an importantrole in inflammatory cytokine production. It functions bybinding to its cell surface receptors IL13Ra1 andIL13Ra2. It has been shown that IL13 blockade by thesoluble muIL13 Ra2 inhibited the signs and symptoms ofasthma in the mouse OVA-induced asthma model.Therefore, inhibition of IL13 signaling has been deemeda potential therapy for asthma. Several in vitro assayshave been set-up to select rat anti mouse antibodies toblock interaction of IL13 with both receptors. An Elisa–based assay has been useful to select rat antibodiesinhibiting binding of muIL-13 to both receptors.However, due to technical difficulties ELISA could onlybe used in selecting antibodies inhibiting binding ofmuIL13Ra1. Here, we describe a Biacore competitionmethod to select rat antibody that inhibit binding toIL13Ra1 and Ra2. Use of this method has been valuablein selection and characterization of rat anti mouseantibodies for in vivo studies.

A105

MICRO-COMPUTED TOMOGRAPHY (mCT) IS APOWERFUL IMAGING TECHNIQUE FOREVALUATING BONE PATHOLOGY INARTHRITIS MODELS

Shaughn Bryant*, Deborah Hyland, Suzanne Mathieu,Michelle Hart, Lisa Olson, Lisa Schopf and AnwarMurtaza

Dept. of Pharmacology, Abbott Bioresearch Center, 100Research Drive, Worcester, MA,01605.

Adjuvant Arthritis (AA) and Collagen-induced arthritis(CIA) are well-established rodent models for rheumatoidarthritis (RA). The disease in these models is initiatedeither by an immunization of an antigen of mycobacterialorigin (AA) or xenogenic type II collagen (CIA). Theseantigens induce a robust pro-inflammatory response,consequently animals develop progressive arthritisinvolving the front and hind limbs with maximal jointswelling most often observed in the affected metatarsal/metacarpal region. One of the hallmarks of AA and CIAmodels is the bone pathology due to an invasiveinflammatory pannus tissue as well as osteoclast mediated

bone resorption. Monitoring these changes in bone arecritical for assessing disease modifying potential of novelanti-rheumatic agents.

Micro-computed tomography (mCT) offers a powerfulimaging technique for evaluating the changes in bone inthese preclinical models. The information captured bymCT can be used to quantitfy bone volume loss, which is ameasure of bone destruction. In addition, more subtlechanges in the bone can be measured by an alternativemCT generated parameter, bone roughness.

We have used the mCT to characterize the changes inbone volume and roughness in the arthritic tarsals. Ourdata for adjuvant or mouse collagen-induced arthritismodels showed a 40-50% reduction in bone volume witha two hundred percent increase in roughness indexcompared to normal age matched controls on days 19and 21 post-immunization for adjuvant arthritis or day 42post-immunization for mouse CIA. Also, a significantincrease in bone roughness was observed in the rat CIAmodel twenty-one days following immunization withbovine collagen. Finally, we have used anti-TNF as acomparator to assess the impact of a disease modifyingagent on bone erosion in our models.

A106

SOFT, TOPICAL IMMUNOSUPPRESSANTSDERIVED FROM CYCLOSPORIN A AND FK-506FOR THE TREATMENT OF ATOPICDERMATITIS

Laurence E. Burgess*, Kevin W. Hunt, Stephen T.Schlachter, David A. Mareska, Robert D. Groneberg,David Chantry, Jed Pheneger, Suzy A. Brown, KenBrameld, Patrice Lee and Kevin Koch

Array BioPharma, 3200 Walnut Street, Boulder, CO 80301

Topical immunosupppressants (pimecrolimus, tacrolimus)are efficacious therapeutics for the treatment of atopicdermatitis. When delivered topically, these calcineurininhibitors offer advantages over topical steroids;however, these marketed drugs have received a blackbox warning because of a potential cancer risk that maybe due to systemic exposure. Accordingly, we havedesigned and discovered a series of “soft” calcineurininhibitors as potentially safer drugs. Soft drugs areengineered, via medicinal chemistry, to be effectiveupon local delivery but, upon systemic exposure, arerapidly metabolized to inactive species resulting in asignificant enhancement in therapeutic index. Relying onolefin metathesis technology, CsA and FK-506 derivativeswere prepared and tested in calcineurin enzyme and IL-2dependent cell assays. Based on the resulting SAR andfurther profiling in hepatocyte and plasma stabilitystudies, ARRY-003 was identified as a potent, softderivative of FK-506. In a swine model of delayed-typehypersensitivity, ARRY-003 displayed excellent efficacyin controlling allergic inflammation, as measured byerythema and induration, when delivered topically.

S94 Inflamm. Res., Supplement 2 (2008)

Page 33: IRA_15th_Conference_21.-24.9.2008

A107

SD0006; A POTENT, SELECTIVE, ANDORALLY-AVAILABLE P38 KINASE INHIBITOR

Barry Burnette*, Shaun Selness, Gail Jungbluth, RaviKurumbail, Stephen Mnich, Richard M. Leimgruber, andJoseph Monahan

Pfizer Global Research and Development, Pfizer,Chesterfield, Missouri

Interest has been high in the pharmaceutical industry inp38a kinase (p38a) as a target for therapeutic drugintervention for diseases such as rheumatoid arthritisbecause of its role in the expression of and the signalingby pro-inflammatory proteins. Here, SD0006 (1-(4-(3-(4-chlorophenyl)-4-(pyrimidin-4-yl)-1H-pyrazol-5-yl) piper-idin-1-yl)-2-hydroxyethanone) was prepared as an inhib-itor of p38a and evaluated both in vitro and in vivo. Invitro, SD0006 was selective for p38a kinase over 50 otherkinases screened, including the g and d p38 isoforms andwith modest selectivity over the b isoform. Crystalstructures with p38a show binding at the ATP site withadditional residue interactions outside the ATP pocketunique to p38a that can confer advantages over otherATP competitive inhibitors. SD0006 suppressed lipopo-lysaccharide (LPS) stimulated expression of pro-inflam-matory cytokines TNFa, IL-1b, and IL-6 with potencies(IC50) <200 nM, and with direct correlation to inhibitionof p38a activity both in cellular models and in vivo,including a clinical trial. SD0006 demonstrated good oralanti-inflammatory efficacy and gave dose-dependentinhibition or arthritis incidence in the mouse CIAmodel equivalent to anti-TNFa treatment.

A108

A ROLE FOR CATHEPSIN K INHIBITORS INRHEUMATOID ARTHRITIS: VEL-0230 PHASE ICLINICAL TRIAL RESULTS

D. Chagnovich*, J. Fellows, R. D. Dyer and M. W. Long

Velcura Therapeutics, Inc., Ann Arbor, MI

VEL-0230* is a potent small molecule inhibitor ofcathepsin K with potential for disease-modifying effectsin RA. A Phase Ia rising-dose safety and tolerance trialindicated both a rapid uptake of the compound (Tmax= 45min) and a rapid reduction of serum collagen type 1 cross-linked C-telopeptide (CTx), a marker of cathepsin K-mediated bone resorption. CTx reduction was equivalentfollowing all doses (150-450 mg), suggesting delivery oflevels of VEL-0230 that achieved or exceeded inhibition-saturating exposures at all doses. CTx was significantlyreduced below that of placebo-treated subjects as early as15 min post-dose and remained significantly belowplacebo control levels through 12 hrs post-dose. An~80% reduction from pre-dose levels of CTx occurred 6hr post-dose. These results demonstrate sustained inhib-ition of bone collagen turnover following single oral dosesof VEL-0230. Plasma concentrations of VEL-0230 weredose-proportional and rapidly cleared, with a T1/2 of ~1 hr.

A single AE of grade 1 dizziness was recorded for oneVEL-0230-dosed patient. Taken together with therecently reported modulation of autoimmune inflamma-tion by this compound**, these results support furtherassessment of VEL-0230 for clinical efficacy in RA andother diseases involving accelerated bone turnover.

*NC-2300; (2S,3S)-3-[[[(1S)-3-methyl-1-[[2-methoxypro-poxy]methyl]butyl]amino]carb-onyl]oxorane-2-carboxy-late, Na+ salt; NC-2300 **Asagiri et al (2008) Science 319,624.

A109

GENOMIC-BASED HIGH THROUGHPUTSCREENING IDENTIFIES SMALL MOLECULESTHAT DIFFERENTIALLY INHIBIT THEANTIVIRAL AND IMMUNOMODULATORYEFFECTS OF IFN-a

Bo Chen*1, Qin Zong2, Ricardo Cibotti1, Chad Morris1,Juana Castaneda2, Brian Naiman1, Derong Liu2, AnnaGlodek2, Gary P. Sims1, Ronald Herbst1, Stephen K.Horrigan2, Peter A. Kiener1, Dan Soppet2, Anthony J.Coyle 1, and Laurent Audoly1,3

1Inflammation and Autoimmunity Group, Research Dept,MedImmune Inc., One Medimmune Way, Gaithersburg,MD 208782 Avalon Pharmaceuticals Inc., Germantown,Germantown, MD 208763 New email address: [email protected]

Multiple lines of evidence suggest that inhibition of TypeI Interferons, including IFN-a, may provide a therapeuticbenefit for autoimmune diseases. Using a chemicalgenomics approach integrated with cellular and in vivoassays, we screened a small compound library to identifymodulators of IFN-a biological effects. A genomicfingerprint was developed from both ex vivo patientgenomic information and in vitro gene modulation fromIFN-a cell-based stimulation. A high throughputgenomic-based screen was then applied to prioritize 268small molecule inhibitors targeting 41 different intra-cellular signaling pathways. Active compounds werefurther profiled for their ability to inhibit the activationand differentiation of human monocytes using disease-related stimuli. Inhibitors targeting NF-kB or JAK/STATsignaling emerged as “dissociated inhibitors” since theydid not modulate IFN-a anti-viral effects against HSV-1but potently inhibited other immune-related functions.This work describes a novel strategy to identify smallmolecule inhibitors for the treatment of autoimmunedisorders.

Inflamm. Res., Supplement 2 (2008) S95

Page 34: IRA_15th_Conference_21.-24.9.2008

A110

THE INDUCTION OF NUCLEAR FACTOR kB INLARYNGEAL CANCER CELLS BY HUMANPAPILLOMAVIRUS-16 ONCOPROTEIN E7 ISASSOCIATED WITH INCREASEDINFLAMMATORY CYTOKINES

Chen G. G.*1, Vlantis A. C.2 , Chun Shan Lo1, JohnsonYip2, Michael C. F. Tong2, van Hasselt C. A.1,2

1Department of Surgery, 2Department ofOtorhinolaryngology Head and Neck Surgery, TheChinese University of Hong Kong, Shatin, New Territories,Hong Kong, China

Laryngeal cancers are frequently infected by high riskHPV-16 and HPV-18. E7 is an oncoprotein of HPV16 andHPV18. Human epithelial cells that express E7 exhibit avariety of growth control defects. The relationshipbetween E7 and Nuclear Factor-kappaB (NF-kB) inlaryngeal cancer is unclear. In this study we haveemployed 2 human laryngeal cancer cell lines (UMSCC12 and UMSCC11) to explore how E7 interacts with NF-kappaB. We have shown that E7 induced increased levelsof NF-kB subunit p65 in the nucleus of laryngeal cancercells. Since NF-kB is a well known survival factor, thisfinding indicates that activation of NF-kB may contributeto cell growth and proliferation of HPV-16-infectedlaryngeal cancer cells. Using immunohistochemicalstaining method, we also showed that p65 was increasedin human laryngeal cancer tissues, compared with non-cancer tissues. We have also observed a significantinfiltration of lymphocytes in the cancer tissues infectedwith HPV-16, suggesting the occurrence of inflammation.Measurement of inflammatory cytokines indicated thatIL-1beta and IL-8 were obviously increased in laryngealcancer cells infected by E7. Accompanying the increase ofIL-1beta and IL-8, the level of p65 was significantlyincreased and the level of p65 was positively associatedwith IL-1beta and IL-8. These changes led to theelevation of the proliferation of tumor cells. Such changescould be prevented when p65 was blocked. In conclusion,The level of NF-kB can be induced by E7, which leads tothe over-production of inflammatory cytokines IL-1betaand IL-8 and subsequently the high proliferation of thetumor cells.

A111

COMPARATIVE EFFECTS OF ANANTI-INFLAMMATORY BLEND ON REDUCINGSKIN IRRITATION CAUSED BY UVB OR ACHEMICAL IRRITANT TO 1%HYDROCORTISONE

Donald Collins*, Neelam Muizzuddin, Chia Chen, DanielMaes

Clinique Laboratories, Melville, NY

It is of considerable interest to develop a topical agent,containing no hy-drocortisone (HC), that can both reducethe onset of chemically or envir-onmentally induced skin

irritation and ameliorate this irritation once it occurs. Ablend of ingredients with anti-inflammatory activities hasbeen developed that outperforms topical 1% HC withregard to UVB induced or Balsam of Peru (BOP)induced skin irritation. This blend contains an inhibitorof histamine release, inhibitors of the PLA2, 5-LO, COX-2, collagenase, elastase and PDE IV enzymes, neutrophilchemotaxis and adhesion blockers, a histamine receptorblocker and an inhibitor of NFkB activation. A cosmeti-cally acceptable oil/water emulsion containing the anti-inflammatory ingredients was prepared and applied tohuman subjects either 20 minutes before exposure to theirritant (UVB or BOP) or after irritant exposure onceerythema was achieved. When applied before the irritant,this blend was able to reduce BOP induced erythema by82% and UVB induced erythema by > 90%. Whenapplied after the irritant, the blend was able to reduceexisting UVB irritation by 22% and existing BOPinduced erythema by 28%.

A112

IMAGING AN INFLAMMATORY RESPONSEUSING 19F MRI

Tracy Monterosso1, Vinod Kaimal1, Patrick McConville1,Robbie B. Mailliard2, Aaron D. Nelson2, Eric T. Ahrens2,Joseph Cornicelli1*

1MRI Preclinical Services, Ann Arbor, MI; 2Celsense,Pittsburgh, PA

Accumulation of mononuclear phagocytes is the hallmarkof chronic inflammation. We present a powerful methodfor noninvasively detecting and quantifying inflammatorycells using 19F MRI. V-SenseM, a 19F-based MRI traceragent is a perfluorocarbon nanoemulsion that is internal-ized by macrophages following intravenous injection. Aninflammatory response was induced in mice using PVAsponges that were soaked in either complete Freund�sadjuvant (CFA) or PBS (control) and implanted subcuta-neously in the dorsal surface of Balb/C mice (day 0). V-Sense (0.2 ml) was injected intravenously at day 4. Micewere imaged on days 5 and 6 at 7T using MRI with a dual19F/1H coil. A 1H anatomical scan was acquired with slicescovering the sponge, liver, spleen and kidneys followed bya 19F scan over the same slices. Significant 19F signal wasobserved surrounding the CFA-soaked sponges, consis-tent with macrophage accumulations but no 19F signalswere detectable in the control PBS-soaked spongeanimals. Necropsies showed that the animals with CFAsponges contained 10–20 fold more inflammatory cellsthan control animals, consistent with the 19F MRI findingsindicating that V-Sense is a specific in vivo biomarker forinflammation that can potentially be used to quantifymacrophage activity.

S96 Inflamm. Res., Supplement 2 (2008)

Page 35: IRA_15th_Conference_21.-24.9.2008

A113

A NOVEL MOUSE MODEL OFMYCOBACTERIUM TUBERCULOSIS-INDUCEDGRANULOMA NECROSIS: IMPLICATIONS FORADJUVANT IMMUNOTHERAPY TARGETINGTH2 CYTOKINES IN TUBERCULOSIS

*Ehlers S.,1 Schreiber T.,2 Thye T.,3 McKenzie A.N.J.,4

Cutler A.,5 Brombacher F.,5 Horstmann R.D.,3 MeyerC.G.,3 Heitmann L.,2 Hçlscher C.2

1Molecular Inflammation Medicine,Christian-Albrechts-University, Kiel, Germany, 2InfectionImmunology, Research Center Borstel, Germany,3Molecular Medicine, Bernhard Nocht Institute forTropical Medicine, Hamburg, Germany, 4MRCLaboratory of Molecular Biology, Cambridge, UnitedKingdom, 5Infectious Disease and Molecular Medicine,UCT, Cape Town, South Africa

TH2 reactions have been implicated in reactivation ofand cavity formation in human tuberculosis (TB). In alarge SNP association study with Ghanaian TB patients,we found a structural variant of the IL-4R-alpha to besignificantly associated with increased lesion and cavitysize. To experimentally address the mechanism of action,IL-13-overexpressing (IL-13tg) mice were aerogenicallyinfected with Mycobacterium tuberculosis (Mtb). Aprofound induction of alternative macrophage activationin IL-13tg mice led to increased bacterial loads onlyduring the chronic stage of infection. Importantly, Mtbinfection in IL-13tg mice, but not in wild type mice,resulted in increased tissue pathology similar to gran-uloma caseation in human TB. IL-4Ralpha-dependentmechanisms represent a novel target for adjuvant immu-notherapy aimed at reducing pathology during chronicand reactivation TB.

A114

P38 MAPK INHIBITOR FOR THE TREATMENTOF RHEUMATOID ARTHRITIS

Michelle Hart*, Shaughn Bryant, Deborah Hyland,Regina Goudreau, Wendy Waegell, Anwar Murtaza, LisaOlson, David Calderwood, and Lisa Schopf

Abbott Bioresearch Center

Rheumatoid arthritis (RA) is an autoimmune diseasecharacterized by chronic synovial inflammation andprogressive destruction of cartilage and bone. p38a is aserine/threonine mitogen-activated protein kinase(MAPK) that is part of a signaling cascade activated bypro-inflammatory stimuli. Inhibition of p38a is expectedto block the production of pro-inflammatory cytokines(e.g. TNF-a, IL-1ß, IL-6, IL-8) and downstream media-tors of inflammation (e.g. COX-2, MMP�s) that areimportant in the pathogenesis of RA. Small moleculep38a inhibitors, from several different chemotypes, havebeen shown to block the production of TNF-a and othercytokines in rodent models. Additionally, thesecompounds have been shown to prevent inflammation

of the joint and bone erosion in various preclinical modelsof arthritis. Using a collagen induced arthritis model inrats, we assessed the impact on inflammation, and bone/cartilage degradation with oral administration of smallmolecule inhibitors of p38a. A serum biomarker ofarthritis, cartilage oligomeric matrix protein (COMP): ameasure of cartilage breakdown that is produced duringinflammation was measured at study termination. Inaddition, we used micro-computed tomographic (mCT)imaging to assess bone destruction in the joints ofarthritic rats. We demonstrate that these different param-eters of measuring bone/cartilage destruction correlatewell with each other and with the traditional paw volumemeasurements. These results suggest that p38a inhibitionmay be an effective disease modifying therapy for thetreatment of RA.

A115

ORAL LACTOFERRIN AND GLYCINE DISPLAYIN VIVO SYNERGISTIC ANTI-INFLAMMATORYACTIVITY

A. Hartog1, 2* and J. Garssen1, 2

1 Danone Research, Centre for Specialized Nutrition,Wageningen, The Netherlands. 2 Department ofPharmacology & Pathophysiology, Utrecht Institute forPharmaceutical Sciences (UIPS), Utrecht University, TheNetherlands

There is a growing awareness of the interaction of foodconstituents with the immune system. The present studiesaim to evaluate immunomodulatory effects of two ofthese nutritional components, i.e. glycine and lactoferrin.Mice orally supplemented with glycine, lactoferrin or acombination, were injected intradermally in the ear, withzymosan. Ear swelling, as a measure for inflammation, aswell as IL-1, TNF-a and IL-6 levels in the ear and thenumber of TNF-a producing spleen cells were analysed.In the collagen induced arthritis (CIA) model mice wereorally supplemented with a combination of glycine andlactoferrin starting after the second collagen booster.Arthritis development was scored and the pro-inflamma-tory cytokine levels in the serum were detected.

Glycine and lactoferrin were able to decrease thezymosan induced inflammatory response locally(decreased ear swelling and pro-inflammatory cytokinelevels) as well as systemically (reduced number of TNF-aproducing spleen cells). Glycine effects (20, 50 and 100mg/mouse/day) were concentration dependent whereasfor lactoferrin only the lowest doses (0.1 and 1 mg/mouse/day) inhibited the inflammatory response significantly.Surprisingly higher doses of lactoferrin (5 and 25 mg/mouse/day) failed to influence the inflammatory reaction.A combination of both nutrients (lactoferrin 0.1mg/mouse/day in combination with glycine 20 or 50 mg/mouse/day) inhibited the zymosan induced ear swellingsynergistically. Additionally, an enhancing effect of bothcomponents was seen on the number of TNF-a producingspleen cells. In the CIA model the combination of glycineand lactoferrin (lactoferrin 0.1mg/mouse/day with glycine20 mg/mouse/day) was able to inhibit arthritis develop-

Inflamm. Res., Supplement 2 (2008) S97

Page 36: IRA_15th_Conference_21.-24.9.2008

ment and to decrease the Il-6 and TNF-a level in theserum significantly.

The present data show anti-inflammatory activity ofglycine and lactoferrin using the zymosan inducedinflammation model. Moreover a combination of bothcomponents demonstrated a synergistic effect on inflam-mation of the skin and a strong anti-inflammatory effectas detected in mouse serum (CIA model).

A116

CHRONIC INFLAMMATION IN ASTHMAAIRWAY REMODELING

Richard Ho*, Ruchi Aggarwal, and Rebecca Baillie

Rosa & Co. LLC, Cupertino, CA

Chronic inflammation has become a dominant factor inour understanding of chronic asthma. While for manyyears the important role that inflammation played inasthma was not recognized and pharmacology recom-mendations were to avoid steroid treatment if at allpossible. Today, clinical authorities recommend inhaledsteroids for all patients with persistent asthma. However,although many studies of chronic asthma patients treatedwith long-term steroids show suppression of inflammationin the lung, patients continue to have exacerbations oftheir disease and relentless decline of FEV1. Thisprogression has been attributed to airway remodeling –structural changes in the airway that obstruct airflow.While experts state that remodeling is triggered bychronic inflammation, the links between inflammationand remodeling and decline in FEV1 are still speculative.We have developed an initial computational model toexplore the physiology of airway remodeling. Thissystems-engineering approach uses physiologicalmodeling and simulation to quantitatively evaluatehypotheses in the progression of the disease. We willpresent the model with details of how it was developedand how it is being applied to drug developmentdecisions.

A117

EFFECTS OF RECOMBINANT HUMAN GROWTHHORMONE ON STAPHYLOCOCCUS AUREUSSEPSIS IN MOUSE

Y. Cao1, C. Yi2, S. H. Mao1, L. L. Ji1, Y. Huang 1*

1 Department of Pathophysiology, West China School ofPreclinical and Forensic Medicine, Sichuan University,Chengdu 610041, China, Fax: ++86 028-85503204, e-mail:[email protected] Cancer Center, Huaxi Hospital, Sichuan University,Chengdu 610041, Sichuan Province, China, e-mail :[email protected]

Objective: To investigate the effects of recombinanthuman growth hormone (rhGH) on Staphylococcusaureus sepsis and explore its possible mechanism.

Methods: Sepsis was mimicked by intraperitoneal (i. p.)injection of Staphylococcus aureus. Male Kun Ming micewere randomly divided into three groups as follows:control group, injected with physiologic saline (i.p.);sepsis group, received a bolus injection of Staphylococcusaureus E311122 (1.75P1012 cfu/L, 40ml/kg, i.p.) followedby intramuscular physiologic saline injection; and rhGHtreatment group, intramuscularly injected with a dose of1.0U/kg rhGH after Staphylococcus aureus challenge.Sepsis group and rhGH treatment group were furtherdivided into 6-, 12- and 24-hr subgroups, respectively. 24hcumulative survival rate, bacteria positive rate of bloodsmear, bacteria count in bacteria plate culture, tumornecrosis factor-a (TNF-a) and interleukin-10 (IL-10)concentrations in plasma, lung injury score, expressionof lung intercellular adhesion molecule-1 (ICAM-1) atlevels of protein and message-RNA (mRNA), nuclearpositive rate of nuclear factor-kappa B (NF-kB) and itsmRNA in the lung were determined.

Results: After Staphylococcus aureus challenge, bacteriapositive rate of blood smear, bacteria count in bacteriaplate culture, plasma TNF-a concentrations, lung injuryscore, and NF-kB and ICAM-1 expression at the levels ofprotein and mRNA in the lung were significantlyelevated, whereas 24h cumulative survival rate andplasma IL-10 levels showed obviously decrease (P <0.05 for all vs. control group). Receiving rhGH treatment,24h cumulative survival rate and plasma IL-10 levels weresignificantly increased, whereas bacteria positive rate ofblood smear, bacteria count in bacteria plate culture, lunginjury score, ICAM-1 and NF-kB protein expression inthe lung were markedly decreased, and the up-regulationof plasma TNF-a was also suppressed. Furthermore,rhGH administration could significantly inhibit the geneexpression of ICAM-1 and NF-kB in the lung (P < 0.05for all vs. sepsis group).

Conclusions: Treatment with rhGH had beneficial effectson Staphylococcus aureus sepsis in mouse, which may beattributed to maintaining a balance of inflammatorycytokines network, reducing the bacterial translocation,inhibiting activation of NF-kB and decreasing ICAM-1expression in lung tissue.

A118

SUPPRESSION OF C - REACTIVE PROTEINAND LIPOPROTEIN LEVELS IN ARTHRITICRATS BY NOVEL GLUCOSAMINE-ANALOG GN1

Huma Jawed*, Shahid I. Awan, Shazia Anjum andShabana U. Simjee

H.E.J. Research Institute of Chemistry, InternationalCenter for Chemical and Biological Sciences, University ofKarachi, Karachi-75270, Pakistan.

The anti-arthritic effects of GN1 (6-hydroxymethyl-3-(1-methylene-allylamino)-tetrahydro-pyran-2, 4, 5-triol) ananalog of glucosamine was investigated on collageninduced arthritis (CIA) in SD rats. Arthritis was inducedin rats by multiple intradermal injections of emulsioncontaining bovine type II collagen in IFA and challenged

S98 Inflamm. Res., Supplement 2 (2008)

Page 37: IRA_15th_Conference_21.-24.9.2008

again with the same antigen preparation 7 days later.Increased hind paw swelling was significantly suppressedwith no further noticeable retardation of body weight inthe groups treated with glucosamine (P < 0.05) and itsanalog GN1 (P < 0.02) as compared to control arthriticrats. In contrast, the animals in the arthritic control groupshowed a gradual decrease in their body weight. Thehistopathological evaluation of isolated knee joints bygrading system, classification of the stages in arthriticlesion development, revealed suppression of the inflam-matory changes in the GN1 treated animals. In addition,both the proinflammatory markers C-reactive protein(CRP) and low-density lipoproteins (LDL) levels werealso found to be significantly decreased in animals treatedwith GN1 (P < 0.03 for CRP and P < 0.05 for LDL ).Thearthritic animals receiving no other treatment or vehicleonly showed a significantly elevated CRP and lipopro-teins levels. These results suggest that GN1 may haveanti-arthritic properties and also act as an anti-inflamma-tory agent.

A119

INHIBITORY EFFECT OF EGCG ON ACUTEPANCREATITIS

Hyo-Jin Ana, Hong-Kun Rima, Jin-Woo Hongb, Hyun-JaJeongc, Seung-Heon Hongd, Jae-Young Uma, Hyung-MinKima,*

aCollege of Oriental Medicine, Institute of OrientalMedicine, Oriental Medical Science Center, Kyung HeeUniversity, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul,Republic of Korea, bDepartment of Cardiovascular andNeurologic Diseases (Stroke Center) College of OrientalMedicine, Kyung-Hee University, Seoul, Republic ofKorea, cBiochip Research Center, Hoseo University, 165,Sechul-ri, Baebang-myun, Asan, Chungnam, Republic ofKorea, dCollege of Pharmacy, Wonkwang University,Iksan, Jeonbuk, Republic of Korea

EGCG has been frequently used as a remedy forinflammatory diseases. The aim of this study was toinvestigate the effect of EGCG on cholecystoki-nin(CCK)-octapeptide-induced acute pancreatitis in rats.EGCG at 10 mg/kg was orally administered, followed by75 mg/kg CCK octapeptide injected subcutaneously threetimes after 1, 3 and 5 h. This whole procedure wasrepeated for 5 d. We determined the pancreatic weight/body weight ratio, the levels of pancreatic HSP60 andHSP72, and the secretion of pro-inflammatory cytokines.Repeated CCK-octapeptide treatment resulted in typicallaboratory and morphological changes of experimentally-induced pancreatitis. EGCG significantly decreased thepancreatic weight/body weight ratio in CCK octapeptide-induced acute pancreatitis. EGCG also increased thepancreatic levels of HSP60 and HSP72. Additionally, thesecretion of IL-6 and TNF-a decreased in the animalstreated with EGCG. EGCG may have a protective effectagainst CCK octapeptide-induced acute pancreatitis

A120

INHIBITORY EFFECT OF INFLAMMATORYMEDIATOR BY OROSTACHYS JAPONICUS INMOUSE PERITONEAL MACROPHAGES

Kyu-Yeop Kima, Hyo-Jin Ana, Hong-Kun Rima, Jin-WooHongb, Hyun-Ja Jeongc, Seung-Heon Hongd, Jae-YoungUma, Hyung-Min Kima,*

aCollege of Oriental Medicine, Institute of OrientalMedicine, Oriental Medical Science Center, Kyung HeeUniversity, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul,Republic of Korea, bDepartment of Cardiovascular andNeurologic Diseases (Stroke Center) College of OrientalMedicine, Kyung-Hee University, Seoul, Republic ofKorea, cBiochip Research Center, Hoseo University, 165,Sechul-ri, Baebang-myun, Asan, Chungnam, Republic ofKorea, dCollege of Pharmacy, Wonkwang University,Iksan, Jeonbuk, Republic of Korea

Orostachys japonicus (OJ) is an herb widely used herbmedicine for the treatment of a variety of pathologies. Inthis study, the effect of OJ on interferon-g (IFN-g) andlipopolysaccharide (LPS)-induced production of nitricoxide (NO), tumor necrosis factor-a (TNF-a), interleukin(IL)-12, and IL-6 were examined using mouse peritonealmacrophages. OJ inhibits IFN-g/LPS-induced NO in adose-dependent manner. The decrease in NO synthesiswas reflected as a decreased amount of inducible NOsynthase protein. We also found that OJ inhibits pro-inflammatory cytokine, IL-12, and TNF-a production.However, OJ doesn�t affect the IL-6 production. Inaddition, OJ inhibited nuclear factor-kB activation andIkB-a degradation. Our study suggests that an importantmolecular mechanism by OJ reduce inflammation, whichmight explain its beneficial effect in the regulation ofinflammatory reactions.

A121

4-ARYL-5-HETEROARYL-2-THIO-SUBSTITUTEDIMIDAZOLES: APPROACH TO P38 MAP KINASEINHIBITOR PRODRUGS

Pierre Koch,a* Jan-Hinrich Guse,b Michael Burnet,b StefanLaufera*

aDepartment of Pharmaceutical/ Medicinal Chemistry,Eberhard-Karls University TMbingen, Auf derMorgenstelle 8, 72076 TMbingen, GermanybSynovo GmbH, Paul Ehrlich Str. 15, D-72076 TMbingen,Germany

The p38a mitogen-activated protein kinase (MAPK) is akey component of the cascade leading to pro-inflamma-tory cytokines like TNF-a and IL-1b. Inhibition of p38MAPK is therefore a promising therapeutic strategy forthe treatment of many inflammatory disorders such asIBD or RA. To date, however, p38 MAPK inhibitorsfailed in late stage clinical development, presumably dueto not tolerable side effects. In order to reduce toxicity,which is mediated by systemic p38 MAPK inhibition, aprodrug concept was established. To this end, new highly

Inflamm. Res., Supplement 2 (2008) S99

Page 38: IRA_15th_Conference_21.-24.9.2008

potent pyridinyl imidazoles were synthesized and linkedto non-antibiotic macrolides. The drug carrier is orallyavailable and concentrated in macrophages and neutro-phils. Using this prodrug system, selected compoundswere investigated for the release of various inflammatoryparameters, like interleukins (IL-2, IL-6), IFN-g, NOproduction and cell proliferation in mouse spleen andperitoneal exudates cells. Most promising candidates andconjugates underwent in vivo studies, e.g. in an acute DSSinduced IBD model and CIA model in mice.

A122

IL-1 DRIVES PATHOGENIC TH17 CELLSDURING SPONTANEOUS ARTHRITIS INIL-1RA-DEFICIENT MICE

Marije I. Koenders*, Isabel Devesa, Renoud J.Marijnissen, Shahla Abdollahi-Roodsaz, Leo A.B.Joosten, Wim B. van den Berg

Rheumatology Research and Advanced Therapeutics,Radboud University Nijmegen Medical Center, TheNetherlands ([email protected])

IL-1Ra-deficient mice spontaneously develop an inflam-matory and destructive arthritis due to unopposed excessIL-1 signaling. In this study, the role of Th17 cells and theeffect of neutralizing IL-17, IL-1, and TNF were inves-tigated. T cells were isolated from IL-1Ra-/- and WTmice, stained for IL-17 and IFNg, and analyzed by FACS.These FACS data showed that an increase of Th17 cellsalready precedes the onset of arthritis in IL-1Ra-/- micewhile % Th1 remains unaffected, and that the percentageof IL-17-positive T cells clearly correlates to the severityof arthritis. IL-1, IL-17, and TNF were highly expressed inthe serum of IL-1Ra-/- mice. Blocking IL-1 or TNFdemonstrated that this IL-1-driven model cannot besuppressed by anti-TNF treatment. Interestingly, thisanti-IL-1 treatment also significantly reduced the %Th17 cells in these arthritic mice. Our blocking studyalso demonstrated that IL-17 contributes to the inflam-mation and bone erosion in this model, and suggests thatIL-1 is the driving force behind the IL-17+ Th17 cells.

A123

IL-17 SYNERGY WITH TNF CAUSES STRIKINGCARTILAGE EROSION IN VIVO

Marije I. Koenders*, Isabel Devesa, Renoud J.Marijnissen, Shahla Abdollahi-Roodsaz, Leo A.B.Joosten, Wim B. van den Berg

Rheumatology Research and Advanced Therapeutics,Radboud University Nijmegen Medical Center, TheNetherlands ([email protected])

To study the combined effects of TNF and IL-17 on jointinflammation and destruction in vivo, mice were intra-articularly injected into the knee joint with adenovirusesencoding for TNF and IL-17.

Local overexpression of either TNF or IL-17 alone causessynovial inflammation and reversible cartilage proteo-glycan depletion. This modest joint pathology was clearlyexaggerated by combining TNF and IL-17. Interestingly,only in the combination group severe chondrocyte deathand cartilage surface erosions were found, indicatingsynergy between TNF and IL-17 on irreversible cartilagedestruction. Overexpression of TNF plus IL-17 signifi-cantly aggravated cartilage VDIPEN expression, amarker for MMP-driven cleavage of aggrecan. QPCRanalysis revealed synergistically elevated mRNA levels ofMMP3 and MMP13 in the cartilage of the TNF+IL-17group. In conclusion, the synergy between TNF and IL-17in vivo results in limited increase in bone erosion, butstriking exaggeration of cartilage erosion, in line withsynergistic upregulation of the erosive enzymes MMP3and MMP13.

A124

FORMYLPEPTIDE RECEPTOR-LIKE-1MEDIATES AMYLOID b-INDUCEDINFLAMMATORY RESPONSE IN ALZHEIMER9SDISEASE

L. Ruan1, J. M. Wang2, Y. Le1,*

1Institute for Nutritional Sciences, SIBS, Chinese Academyof Science, Shanghai, China; 2Laboratory of MolecularImmunoregulation, NCI-Frederick, Frederick, Maryland,USA

Amyloid beta (Ab) is a major contributor to the patho-genesis of Alzheimer�s disease (AD). Accumulatingevidence indicates that Ab causes neuronal damage byactivating glial cells that accumulate in and around senileplaques. Our previous studies revealed that formylpeptidereceptor-like-1 (FPRL1) mediated the chemotactic effectof Ab42 on mononuclear phagocytes (monocytes andmicroglia), and elevated FPRL1 gene expression wasdetected in CD11b-positive mononuclear phagocytes thatinfiltrate the plaques in brain tissues of the AD patients.In this study, we examined the involvement of FPR2, themouse homologue of human FPRL1, in Ab42-inducedinflammatory response both in vitro and in vivo. Wefound that lowering the expression of FPR2 usinglentiviral vector expressing siRNA targeting FPR2 abol-ished Ab42-induced IL-1b, IL-6 and MCP-1 expression atmRNA level and protein levels in mouse primary micro-glia and astrocytes, as well as iNOS and TNF-a mRNAexpression in mouse primary astrocytes. Furthermore,lentivirus-mediated FPR2 RNA interference in hippo-campus of mice inhibited Ab42-induced microglia andastrocyte activation as well as TNF-a and IL-6 up-regulation. Thus, FPRL1 may mediate inflammation seenin AD and is a potential target for developing therapeuticagents.

S100 Inflamm. Res., Supplement 2 (2008)

Page 39: IRA_15th_Conference_21.-24.9.2008

A125

OPTIMIZATION OF A QUINOLONE CLASS OFDISSOCIATED GLUCOCORTICOID MIMETICS

T. W. Lee,* J. Regan, Y. Bekkali, J. Bentzien, A. J.Kulkulka, J. Kahn, L. Zuvela-Jelaska, D. Souza, G.Nabozny, D. S. Thomson

Departments of Medicinal Chemistry and Immunology &Inflammation, Boehringer Ingelheim Pharmaceuticals,Inc. Ridgefield, CT 06877

Glucocorticoids (GCs) and their derivatives have foundwide use in the treatment of many inflammatory diseases.Research efforts are now focused on “dissociated” GCderivatives or mimetics that exhibit reduced side effectprofiles versus traditional GCs while maintaining thepotent anti-inflammatory activities. Recently, wedisclosed a quinolone class of dissociated GC mimeticswith potent antiflammatory activities in cellular and invivo assays. These earlier compounds with poor PK werenot suitable for testing in a chronic disease relevantmodel (CIA). Further optimization led to BI-306 (3-chloro-1-{4-[5-(2,6-dimethyl-pyridin-4-yl)-2,3-dihydro-benzofuran-7-yl]-2-hydroxy-4-methyl-2-trifluoromethyl-pentyl}-1H-quinolin-4-one) with the following improvedprofile: (a) potent and selective over other nuclearreceptors, (b) dissociated in cellular systems, (c) improvedplasma level after oral dosing, (d) potent inhibition ofLPS-induced TNFa production, and (e) efficacious in anAb CIA model.

A126

INFLAMMATORY SIGNALING PROCESSES ANDCARDIOVASCULAR COMPLICATION

Li L.*, Wang D., Fasciano S., Gan L., Maitra U.

Virginia Tech, Blacksburg, VA

Inflammatory signaling processes are critically involvedin the pathogenesis and resolution of various inflamma-tory diseases such as atherosclerosis and hypertension.The interleukin-1 receptor associated kinase 1 (IRAK-1)is a critical modulator regulating the inflammatorysignaling processes. We have demonstrated that IRAK-1is a kinase responsible for the phosphorylation andinactivation of the Nuclear Factor of Activated T-cell(NFAT). Expression of IRAK-1 suppressed NFATreporter activity. Correspondingly, the levels of bothnuclear NFATc1 and NFATc4 were constitutivelyelevated in IRAK-1-/- cells. Furthermore, the phosphor-ylation of NFATc4 at the S168PS170P site was significantlydiminished in IRAK-1-/- cells. Mechanistically, weobserved that IRAK-1 interacted with NFATc4 via theC-terminus of IRAK-1 and the N-terminal NHR regionof NFATc4. IRAK-1 mutants that ablated either itskinase activity or its interaction with NFATc4 failed tosuppress NFAT reporter activity. The expression level ofCOX-2, which is under the control of NFAT, was elevatedin IRAK-1-/- cells. Functionally, ApoE-/-/IRAK-1-/- micewere protected from high-fat-diet induced hypertension

and atherosclerosis. Taken together, our findings revealthat IRAK-1 serves as a novel target for treatingcardiovascular diseases.

A127

THE GOLGI-ASSOCIATED PROTEIN P115MEDIATES THE SECRETION OF MACROPHAGEMIGRATION INHIBITORY FACTOR (MIF)

Melanie Merk*, John Baugh, Swen Zierow, Seung JoonLee, Paula Kavathas, Richard Bucala

Yale University School of Medicine, New Haven, CT,06520

The leaderless pro-inflammatory cytokine MIF is secretedfrom cells by an unconventional export pathway. Therelease of MIF plays an important role in diverseinflammatory diseases. We identified the Golgicomplex-associated protein, p115, as an intracellularbinding partner for MIF. MIF localizes with p115 in thecytoplasm and the stimulated secretion of MIF results inthe accumulation of both proteins in supernatants, whichis consistent with MIF release from cells in conjunctionwith p115. The depletion of p115 from monocytes/macrophages decreases the release of MIF but notother cytokines following inflammatory stimulation orintracellular bacterial infection. Notably, the small mole-cule MIF inhibitor, 4-iodo-6-phenylpyrimidine, inhibitsMIF secretion by targeting the interaction between MIFand p115. These data reveal p115 to be a criticalintermediary component in the regulated secretion ofMIF from monocytes/macrophages.

A128

GABA(A)-MEDIATED ALTERATION OFAUTOIMMUNE-MEDIATED INFLAMMATIONUSING THE NATURAL PLANT PRODUCT,HONOKIOL

Melissa E. Munroe*1 and Gail A. Bishop1, 2, 3

1University of Iowa Dept. of Microbiology, 2Dept. ofInternal Medicine, 3VAMC

Honokiol (HNK) is a natural, purified, product of theleaves and root stems of the Magnolia plant that has longbeen used in traditional Asian medicine withoutdangerous side effects. To determine if HNK has anti-inflammatory properties, we tested its effects in a mousemodel of inflammatory rheumatoid arthritis (RA). Ourinitial studies show that both in vivo inflammation anddisease progression and in vitro pro-inflammatory cyto-kine production and cellular activation are markedlyinhibited, without increased cell death or in vivo toxicity.HNK also inhibits signaling via CD40 in B cells, animmune receptor that has been implicated as playing arole in RA, as well as an EBV-encoded CD40 mimiccalled LMP1, that has been implicated in exacerbating

Inflamm. Res., Supplement 2 (2008) S101

Page 40: IRA_15th_Conference_21.-24.9.2008

autoimmune disease. Furthermore, the anti-inflammatoryeffects of HNK could be reversed using inhibitors of theneurotransmitter GABA(A), a previously reported targetfor HNK interaction. These findings are particularlyexciting and suggest that the nontoxic anti-inflammatoryproperties of HNK could prove more effective thantreatments targeting a single cytokine or receptor.

A129

HUMAN APOLIPOPROTEIN C1 TRANSGENICMICE: A UNIQUE NOVEL MODEL OF ATOPICDERMATITIS

Lex Nagelkerken*, Perry Verzaal, Richard Verbeek, TonnyLagerweij, Carla Persoon-Deen, Louis Havekes andArnold P. Oranje

TNO Quality of Life, Inflammatory & DegenerativeDiseases, Leiden, and Erasmus Medical Center,Rotterdam, The Netherlands.

Mice with a chronic overexpression of human apolipo-protein C1 in liver and skin display increased levels ofcholesterol and triglycerides, and spontaneously developclinical symptoms of atopic dermatitis (AD). These miceshow increased trans-epidermal water loss suggestive foran impaired skin-barrier function. Development of AD inthese mice is associated with increased pruritus. Histo-logical analysis shows hyperplasia of both the epidermisand dermis and increased numbers of CD4+ T cells,eosinophils and IgE+ mast cells in the dermis. Serumlevels of IgE are increased as well. Development of ADin this model was found to be sensitive to topicaltreatment with triamcinolone-acetonide, fluticasone-isoproprionate or tacrolimus. Moreover, oral treatmentwith dexamethasone successfully inhibits several aspectsof disease in this model. Therefore, this novel animalmodel may help to gain new insight into the pathogenesisand be of great value to develop novel therapeuticstrategies for the management of AD.

A130

MODULATION OF INFLAMMATION IN TWOHUMANIZED MOUSE MODELS OF PSORIASIS

Perry Verzaal, Monique Smits, Carla Persoon-Deen, Koenvan der Mark, Nicole Worms, Arianne Plomp and LexNagelkerken*

TNO Quality of Life, Inflammatory & DegenerativeDiseases, Leiden, The Netherlands.

Several mechanisms involved in the development andprogression of psoriasis are considered as potentialtherapeutic targets for biologicals. To ensure that suchreagents properly predict efficacy in the clinic it is of highimportance to evaluate those in a humanized model ofpsoriasis. We here describe two humanized mouse modelsof psoriasis by grafting either non-lesional or lesional skinfrom psoriasis vulgaris patients onto immunodeficientmice. In the non-lesional skin graft model a psoriatic

process is induced by intradermal injection of SEB-activated autologous T cells. In both models a psoriaticprocess is demonstrated by assessment of epidermalhyperplasia, and proliferation (Ki67) and differentiation(CK16) of keratinocytes, in conjunction with the involve-ment of innate cells, activated T cells and cytokines likeTNF-a. Either model was shown to be sensitive to a widerange of therapeutics, including anti-TNF-a and anti-IL-23. Therefore, these two humanized mouse modelsrepresent a powerful tool for the identification orvalidation of potential therapeutics in psoriasis.

A131

EFFICACY OF PROBIOTICS IN TNBS-INDUCEDCOLITIS

Tonny Lagerweij, Frans Tielen, Nicole Worms, CarlaPersoon-Deen, Maria Stolaki, Sonia Pavan, & LexNagelkerken*

TNO Quality of Life, Inflammatory & DegenerativeDiseases, Leiden, The Netherlands.

Defects related to epithelial barrier function, NOD2,altered production of IL-12 and IL-23 by dendritic cells,strong Th17 responses and impaired regulatory T cellshave been implicated in Inflammatory Bowel Disease. Weevaluated the sensitivity of TNBS-induced colitis inBALB/c mice with respect to immunosuppressive drugsand probiotics. Systemic administration of corticosteroids,CsA, IL-10 or Etanercept were mostly ineffective, or evenresulted in increased mortality. Rectally instilled budeso-nide resulted in up to 50 % inhibition of colitis asmeasured by effects on colon damage and cellularinfiltration. Of note, treatment with L.plantarum orVSL#3 had substantial effects, evident from a decreasein infiltrating CD4+, CD8+ and CD11b+ cells in thecolon. Interestingly, these observations were associatedwith decreased serum levels of IL-17, IFN-g, IL-1b andMIP-1a. Such effects were also found in mice treated withIL-10 or Etanercept, indicating – in view of the increasedmortality - that immunosuppression may not always bethe right strategy of treatment in experimental colitis. Ourdata are in favor of treatment with probiotics as anapproach to restore intestinal immune homeostasis.

A132

PREDICTING EFFICACY AND SIDE EFFECTS OFTHE P38MAP KINASE INHIBITOR CLASS USINGBIOMAP: PRIMARY HUMAN CELL-BASEDSYSTEMS

A. O�Mahony*, S. Privat, D. Nguyen, E. Rosler, J.Melrose, E.L. Berg and E.J. Kunkel

BioSeek, Inc., Burlingame, CA 94010

Inhibitors of p38 are safe and strongly anti-inflammatoryin preclinical animal models, but have reported dose-limiting liver and skin toxicity in human clinical trials.BioSeek has pioneered the development of in vitro

S102 Inflamm. Res., Supplement 2 (2008)

Page 41: IRA_15th_Conference_21.-24.9.2008

human cell-based BioMAPM Systems that incorporate thecomplexity of environmental factors observed in diseasedtissues in vivo. Analysis of multiple experimental andclinical p38 inhibitors in BioMAPM identified class-specific biomarker and pathway modulation informationthat is predictive for both efficacy (monocyte/macro-phage-driven inflammation) and side effects (liver andskin tissue stress responses) with strong correlation toclinical data. In particular, the liver and skin stressresponses seen with p38 inhibitors in BioMAPM areshared with other therapies known to cause suchresponses (i.e. EGF pathway inhibitors). Inhibitors ofdownstream p38 targets (e.g. MK-2 or MNK1) havereduced anti-inflammatory activity, but also induce less ofa stress response. These findings demonstrate howBioMAPM human disease models can drive the discoveryand optimization of safe and efficacious therapeutics.

A133

HISTAMINE IS NOT RELEASED IN ACUTETHERMAL INJURY IN HUMAN SKIN IN VIVO :A MICRODIALYSIS STUDY

L.J. Petersen*, J.L. Pedersen, P.S. Skov, H.J. Nielsen, andH. Kehlet

The Department of Clinical Physiology, Viborg Hospital,Viborg, Department of Surgical Gastroenterology,Hvidovre Hospital, Hvidovre, and The ReferenceLaboratory, National University Hospital, Copenhagen,Denmark.

Background Animal models have shown histamine to bereleased from the skin during the acute phase of athermal injury. The role of histamine during the earlyphase of thermal injuries in humans remains unclear.

Purpose The objectives of this trial were to studyhistamine release in human skin during the acute phaseof a standardized thermal injury in healthy volunteers.

Methods Histamine concentrations in human skin weremeasured by skin microdialysis technique. Microdialysisfibers were inserted into the dermis in calf skin in malehealthy volunteers. A standardized thermal injury waselicited by a heating thermode. Histamine in dialysatewas analyzed for up to 120 min.

Results In separate investigations, histamine was analyzedin 2-min samples over 20 min (n=6) and at 10-minintervals over 120 min (n=8) after the injury. Histaminelevels at baseline and in most port-injury samples were ator below the quantification limit of the spectroflouro-metric analysis. Confirmatory analysis using a sensitiveradioimmunoassay showed no significant histaminerelease during the first 60 min after a thermal injuryusing a (baseline histamine 11.6 � 1.8 nM versus 14.8 �1.8 nM post injury).

Conclusions Histamine is not released in human skinduring the acute phase of a thermal injury.

A134

EFFECTS OF A SELECTIVE, POTENT P38INHIBITOR IN IN VIVO MODELS OFRHEUMATOID ARTHRITIS

Jed Pheneger*, Dale Wright, Alison Bendele, LynelleLopez, Kevin Koch, Jim Winkler, Patrice Lee

Array BioPharma and Boulder BioPath, Boulder CO,USA

ARRY-371797 (ARRY-797; (N-substituted-5-(2,4-difluor-ophenoxy)-1-isobutyl-1H-indazole-6-carboxamide). ) is aselective, potent inhibitor of the p38a enzyme(IC50<5nM). This activity is maintained in human wholeblood cytokine assays (IC50<2nM). Here we show theeffects of ARRY-797 in two rat models of rheumatoidarthritis (CIA and AIA) when given alone or incombination with methotrexate. In CIA, ARRY-797 (3,10, or 30 mg/kg, BID,PO) dosed as a single agent (d10-16)inhibited paw diameter increases with an ED50 of ~5 mg/kg and histological lesions with an ED50 of ~3 mg/kg. InAIA, ARRY-797 (3, 10, or 30 mg/kg, PO, BID) was dosedas a single agent (d0-16) or with methotrexate (MTX;0.05mg/kg QD, PO, d0-16). ARRY-797 alone inhibited pawdiameter (~40%) at 30 mg/kg and was very effective inthe inhibition of bone resorption (ED50 of <3 mg/kg).ARRY-797 with MTX resulted in additive inhibition ofpaw swelling. We have shown the potent p38 inhibitorARRY-797 has excellent anti-inflammatory and boneprotective effects in CIA and potent inhibitory effects onbone resorption in AIA. ARRY-797 can be combinedwith MTX to produce at least additive effects on in-lifeand histological endpoints. ARRY-797 is in clinical trialsto study its safety and efficacy in patients with pain andother inflammatory disease.

A135

A GLOBAL GENE EXPRESSION PROFILINGANALYSIS TO STUDY THE ROLE OFENDOTHELIAL CD40 DURING INFLAMMATION

Pluvinet R.*, Olivar R., Krupinski J., Sumoy L. and AranJ.M.

CGMM- IDIBELL, Hospitalet de Llobregat, Barcelona,Spain.

The CD40-CD154 interaction plays a key role in theimmune-inflammatory response triggered by endothelialcell (EC)-T cell communication. To study the involve-ment of CD40 in EC activation we have carried out aglobal gene expression profiling analysis of ECs inter-acting with (CD154+) T cells. We have assessed theconsequences of blocking CD40 signaling with a specifichuman anti-CD40 siRNA in a time course experimentand have observed an extensive transcriptional responseafter CD40-CD154 engagement in ECs. There is an earlyup-regulation of the major pro-inflammatory NF-kB andMAPK/SAPK pathways and their associated transcrip-tion factors. Moreover, CD40-mediated up-regulation ofthe viral immune surveillance system, notably TLR3,

Inflamm. Res., Supplement 2 (2008) S103

Page 42: IRA_15th_Conference_21.-24.9.2008

IFIH1, RIG-I and RNASEL, establishes a reverse linkfrom adaptive to innate immunity. This suggests thatCD40 triggers the antiviral innate immune response inECs when challenged with activated T cells. In summary,we have identified novel genes and signaling pathwaysinvolved in the induction of the inflammatory responsethrough CD40 activation in the endothelium.

A136

DIPEPTIDYL PEPTIDASE I MEDIATESCIGARETTE SMOKE-INDUCED PULMONARYINFLAMMATION AND ALVEOLARDESTRUCTION

Patricia L. Podolin*, Joseph P. Foley, Brian J. Bolognese,William E. Wixted, Jen P. Kou, Elizabeth A.Capper-Spudich, and Michael S. McQueney

GlaxoSmithKline, King of Prussia, PA

Based on evidence that a protease-antiprotease imbal-ance is pivotal in the development of chronic obstructivepulmonary disease (COPD), we hypothesized that dipep-tidyl peptidase I (DPPI), the physiological activator

of neutrophil elastase (NE), may contribute to theetiology of COPD. DPPI gene-deficient (DPPI-/-) miceexposed to 4% mainstream cigarette smoke (CS) for 2weeks exhibited reduced numbers of BAL fluid neutro-phils, macrophages, and lymphocytes, as well as decreasedlevels of KC, MCP-1 and IL-12p40, compared to DPPI+/+

and DPPI+/- mice. Following 15 weeks

of CS exposure, DPPI-/- mice exhibited reduced numbersof BAL fluid neutrophils and macrophages, but notlymphocytes, compared to DPPI+/+ and DPPI+/- mice.DPPI-/- BAL fluid levels of KC and MCP-1 were alsodecreased, while IL-12p40 levels were not. Importantly,DPPI-/- mice were completely protected from CS-inducedairspace enlargement. In a second model of COPD,DPPI+/+ mice exposed to ozone (3 ppm for 3 hours) twiceweekly for 6 weeks exhibited significant airspace enlarge-ment, while DPPI-/- mice were not different from sham-exposed mice. These results suggest that DPPI inhibitionoffers a promising approach to the treatment of COPD.

A137

THE INVOLVEMENT OF NITRIC OXIDE IN THEPERIPHERAL ANTINOCICEPTIVE EFFECTS OFOPIOIDS DURING CHRONIC INFLAMMATORYPAIN

I. Torres, S. LeNnez and O. Pol*

Laboratori de Neurofarmacologia Molecular, Institut deRecerca, Hospital de la Sta Creu i Sant Pau & Institut deNeurociOncies, Universitat AutPnoma de Barcelona,Barcelona, Spain.

We investigated using inducible nitric oxide synthase(NOS2) knockout mice the role of nitric oxide in the local

antinociceptive effects of m and d-opioid receptor agonistsduring CFA-induced peripheral chronic inflammatorypain. The presence of paw inflammation, mechanicalallodynia and thermal hyperalgesia induced by CFA wereassessed by measuring paw diameter and using the vonFrey filaments and plantar tests, respectively. NOS2knockout mice exhibited reduced paw edema and dimin-ished thermal hyperalgesia after CFA as compared to WTmice. The subplantar administration of morphine (m-agonist) or [D-Pen2,5]-enkephalin (d-agonist) whichcompletely reversed the thermal hyperalgesia inducedby chronic inflammatory pain in WT mice was completelyineffective in NOS2 knockout mice. These results indicatethat nitric oxide derived from NOS2 participates in pawedema and thermal hyperalgesia induced by CFA as wellas in the antinociceptive effects of m- and d-opioidreceptors during chronic inflammatory pain. Supportedby FIS (05/1604) & FundaciQ MaratQ TV3 (07/0810),Spain.

A138

CAMPYLOBACTER JEJUNI-INDUCEDACTIVATION OF MURINE DENDRITIC CELLSINVOLVES COOPERATIVE SIGNALINGTHROUGH MYD88 AND TRIF

V.A.K. Rathinam*, D.M. Appledorn, K.A. Hoag, A.Amalfitano and L.S. Mansfield

Michigan State University, East Lansing, MI-48824.

Campylobacter jejuni (Cj) is a frequent cause of humanenteritis that resembles inflammatory bowel disease. Weshowed that Cj induces activation of bone marrow-derived dendritic cells (BM-DCs). Toll-like receptors(TLRs) of DCs induce immune responses to pathogensthrough adapters such as MyD88 and TRIF. We hypothe-sized that Cj-induced inflammatory activation of BM-DCs is mediated by TLR2 and TLR4 and that MyD88 orTRIF signaling is necessary for activation. WT, MyD88-/-,TRIF-/-, TLR4-/- or TLR2-/- BM-DCs were exposed to Cjand immune responses were assessed. Up-regulation ofMHC-II after Cj challenge was significantly impaired byMyD88-, TRIF-, TLR4- or TLR2-deficiency. Increase inexpression levels of CD80, CD86 and CD40 and secretionof IL-12, IL-6 and TNF-a following Cj challenge wassignificantly inhibited in MyD88-/-, TRIF-/-, TLR4-/- andTLR2-/- DCs compared to WT DCs. However, themagnitude of inhibition was greater in MyD88-/-, TRIF-/-

and TLR4-/- DCs than in TLR2-/- DCs. Our results showfor the first time that cooperative signaling throughTLR4-MyD88 and TLR4-TRIF axes represents a novelmechanism mediating Cj-induced maturation and inflam-matory responses of DCs. (Funded by NIH contract NO1-AI-30058 and grant K26 RR023080-01).

S104 Inflamm. Res., Supplement 2 (2008)

Page 43: IRA_15th_Conference_21.-24.9.2008

A139

AN2898: A NOVEL ANTI-INFLAMMATORYCOMPOUND THAT INHIBITSPHOSPHODIESTERASE 4 AND 7 ENZYMEACTIVITY AND IL-12 AND IL-23 RELEASE

Virginia Sanders*, Fernando Rock, Richard Kimura,Joanna Antunes, Yvonne Freund, MRK Alley, YasheenZhou, Tsutomu Akama, Kirk Maples, Jake Plattner

Anacor Pharmaceuticals, Inc., 1020 East Meadow Circle,Palo Alto, CA 94303, USA.

AN2898 (5-(3,4-dicyanophenoxy)-1-hydroxy-1,3-dihydro-2,1-benzoxaborole) is a dual-inhibitor of PDE4 andPDE7 enzymes. Additionally, AN2898 inhibits IL-12and IL-23 release from PBMCs. This new small moleculecompound is currently in preclinical development forpsoriasis and atopic dermatitis, common skin diseases thatare characterized by chronic inflammation. AN2898 IC50

values for inhibition of PDE4 and PDE7 activity is 0.06and 0.21mM respectively. AN2898 is a competitive,reversible inhibitor of PDE4 with a Ki of 65 nM. TheAN2898-PDE4B2 catalytic domain co-crystal structureshows that AN2898 binds directly to the metal ions andwater molecule in the active site. This binding config-uration is unique compared to classical PDE4 inhibitors(i.e. rolipram). AN2898 has equal activity against the fourPDE4 subtypes and does not significantly inhibit PDE 1,2, 3, 5, or 6. Unlike classical PDE4 inhibitors, AN2898 hasadditional activity and inhibits IL-12 (IC50 = 0.016 mM)and IL-23 (IC50 = 1.1 mM). These are important cytokinesin the treatment of psoriasis, and are not affected byPDE4 inhibition. Like other PDE4 inhibitors, AN2898inhibits the release of TNF a, IL-2, IFN g, IL-5, and IL-10in the low to mid-nanomolar range, and does not inhibitIL-1 b, IL-6 and IL-8. AN2898 after topical administra-tion demonstrates significant in vivo activity. AN2898 hasmuch greater efficacy compared to rolipram in a PMAinduced ear edema model. Additionally, AN2898 hasanti-inflammatory activity in the oxazolone model (amodel of allergic contact dermatitis). In conclusion,AN2898 shows excellent activity against cytokines asso-ciated with psoriasis and atopic dermatitis.

A140

DISCOVERY OF3-{3-(2-PIPERIDINYLETHOXY)PHE-NYL}-5-(1H-1,2,4-TRIAZOL-3-YL)-1H-INDAZOLE(CC-401), A POTENT JNK INHIBITOR

Y. Satoh,* S. Sakata, A. Kois, V. Plantevin, K.Sahasrabudhe, Q. Chao, C. Buhr, W. Lew, G. Shevlin, R.Albers, L. Nadolny, N. D�Sidocky, J. Sapienza, R. Ferri, M.McCarrick, A. Motiwala, J. Muir, C. Grimshaw, W. Xu,L.Xu, O. Khatchenko, S. Pai, M. A. Shirley, E. O�Leary,H. Raymon, P. Omholt, J. Leisten, J. Wright, S. Bhagwat,A. Manning, A. Lewis, G. Friedman, T. Uehara, G. Bilter,J. Westwick, D. Brenner, and B. Bennett

Celgene, 4550 Towne Centre Court, San Diego, CA 92121,USA

Activation of Jun N-terminal kinases (JNK�s) inischemia-reperfusion (IR) injury is well documented.Through the template modification efforts on a screeninghit, anthrapyrazolone (SP600125), we identified a potent,indazole-based JNK inhibitor, CC-401, which inhibitedJNK�s (JNK2; Ki = 24 nM), blocked PMA/PHA inducedIL-2 production in Jurkat T-cells (IC50 = 0.8 mM), andprevented deaths in a rat liver IR injury model (84%survival at 10 mg/kg iv CC-401 vs. 0% in control). CC-401 was investigated in a number of normal volunteersand patients in Phase 1 trials to monitor pharmacokineticsand proof of JNK inhibitory activity.

A141

RESOLVIN E1 (RVE1) INHIBITS INFLAMMATIONIN ACUTE AND CHRONIC MURINE MODELSOF COLITIS

Savinainen A.*, Crandall T., Lin M., Wu L., Picarella D.

Resolvyx Pharmaceuticals Inc, Bedford MA

Resolvins are a recently discovered family of naturally-occurring, small molecule lipid mediators that act toresolve inflammation, protect healthy tissue and restoreimmune homeostasis. Resolvins show highly potentefficacy in rodent models of rheumatoid arthritis, asthmaand other inflammatory diseases. These molecules mayrepresent a novel class of therapeutics for the treatmentof a wide range of inflammatory diseases. In these studies,the objective was to evaluate the therapeutic effect ofRvE1 in acute and chronic models of inflammatory boweldisease (IBD), including oral consumption of dextransodium sulfate (DSS) and adoptive transfer of CD45RB-hi CD4+cells T cells into immunodeficient recipients.Therapeutic endpoints in these studies included bodyweight, colon length, colon edema, myeloperoxidase(MPO) activity, and histological scores. The results fromthese studies suggest that RvE1 effectively modulatesdisease pathology and could have therapeutic potential inthe treatment of IBD.

A142

THE POLY-ANIONIC SUGAR SUCROSEOCTA-SULPHATE IS AN ORALANTI-RHEUMATIC AND ANTI-EROSIVEMETABOLITE OF SUCRALFATE

Jones R., Mancini J., Lees M., Roitt I. , Burnet M.†,Seed M.*

William Harvey Res. Inst. , Queen Mary�s Barts & LondonMed. School, London EC1M 6BQ, UK. † Synovo GmbH,Tubingen, D72076 Germany.

We have reported that polyanionic diglucopyranosyl-amines are antirheumatic in vivo, and here the disac-charide analogue sucrose octasulphate (SOS). Antigeninduced arthritis (AIA) C57bl mice sensitized to methy-lated BSA challenged ia, and joint dia. Assessed at 24hrs.Murine (dba-1, MCIA) and rat (Lewis, RCIA) collagen

Inflamm. Res., Supplement 2 (2008) S105

Page 44: IRA_15th_Conference_21.-24.9.2008

arthritis was induced, scored and paw volume (plethys-mometry) measured. CT images of fixed paws wereacquired (Siemens Microcat II instrument), isosurfaceplots obtained with reference to an untreated control, andscored. 100mg/kg p.o. sucralfate inhibited AIA, and thisreversed by lansoprazole. SOS (100, 30, 10mg/kg p.o.)dose relatedly inhibited AIA. RCIA was inhibited whendosed p.o. from day 0-11, and MCIA dosed p.o. prophy-lacticaly. An MCIA bone erosion score was developed,and erosion scores reduced from 12.0�3.4 to 6.5�2.7(p<0.07, n=9 & 10). Dexamethasone reduced scores from10.4�2.9 to 2.4�0.8 (p<0.01, n=1- & 12). Orally activepolyanionic drugs may provide a novel approach to oralanti-rheumatic anti-erosive therapy.

A143

PRECONDITIONING LYMPHOCYTES WITH P38MAPK INHIBITORS, AND NOT ACCESSORYCELLS, PREVENTS CON-A-INDUCEDLYMPHOCYTE RESPONSES

V Moradi, E Johnson, L Dugo, V Holan, M Burnet, SLaufer, M Seed*

KINACEPT Team, William Harvey Res. Inst. LondonEC1M6BQ.

We have shown DC-T cell MLR requires active T cell p38MAPK before stimulation, but not in DCs. We here assesswhether this is specific for antigen dependent responses.Balb/c mouse spleen cells were stimulated with 2.5mg/mlCon-A in the presence or absence of 3mM SB203580 (4-(4-Fluorophenyl)-2-(4- ethylsulfinylphenyl)- 5- (4-pyridyl) 1H-imidaz- ole, SB) or 3mM ML3403 ((RS)-{4-[5-(4-Fluorophenyl)-2-methylsulfan- yl-3H-imidazol-4-yl]pyridine-2-yl}-(1-phenylethyl)amine, ML) and incu-bated for 72 hours. Accessory cells (AC) were separatedfrom LØ by adherence to plastic and washed 3 times.Separated cells were pre-conditioned with drugs for 2hours, washed 3 times, resuspended (1x106/ mL), mixedand stimulated with Con-A. Con-A induced LØ prolifer-ation was dependent on ACs. SB and ML both inhibitedwhole spleen cell proliferation. 2hr preconditioning ofACs with SB or ML had no effect, whilst preconditioningof LØ inhibited proliferation. Thus both SB and ML areeffective after washout. LØ, not AC, require functionalp38 prior to Con-A stimulation in order to proliferate.P38 inhibition conditions LØ responsiveness to AC andDCs.

A144

PREVENTIVE ROLE OF ZH-67-2-1 INADJUVANT-INDUCED ARTHRITIS IN RATS

S. Uzair Shah*, Huma Jawed, Zahid Hussain, M. RazaShah and Shabana U. Simjee

H.E.J. Research Institute of Chemistry, InternationalCenter for Chemical and Biological Sciences, University ofKarachi, Karachi-75270, Pakistan.

In the present study, the anti-arthritic and anti-oxidativeeffects of the synthetic compound ZH-67-2-1 (6-nitro-1,3-benzodioxane, Fig. 1), was evaluated in the adjuvant-induced arthritic (AIA) rats. It was observed that theincreased hind paw swelling was significantly suppressed(p <0.007) in the ZH-67-2-1 (20 mg/kg, i.p.) treatedarthritic rats as compared to arthritic control rats. Bodyweights were also measured to monitor the progression ofdisease and systemic anti-arthritic effects of the testcompound used in this study. Our results show that ZH-67-2-1 not only inhibited the macroscopic changes such aserythema and swelling of limbs but also exhibited reversalof nociception as measured by traction test. Moreover,the body weight reduction which was observed in arthriticand saline treated arthritic control rats was not observedin the arthritic rats treated with ZH-67-2-1. The anti-oxidative activity of ZH-67-2-1 was measured in theserum of all treated and untreated arthritic and non-arthritic rats. The ZH-67-2-1 treatment was observed tosignificantly suppress both the nitric oxide (NO) (p <0.014) and peroxide levels (p < 0.00) as compared to thearthritic control group. Based on these observations, wesuggest that one of the possible mechanism through whichZH-67-2-1 exerts its anti-arthritic effect is through its freeradicals scavenging activity.

A145

DESIGN OF SMALL MOLECULE INHIBITORSFOR INFLAMMATORY BOWEL DISEASEMECHANISM: INFLAMMATORY/IMMUNEMECHANISMS

G.V.R. Sharma*, N. Sukunath, Uma Ramachandran,Sriram Rajagopal, K.V. Sanjay, S. Thirunavakkarasu, D.Bhakiaraj

Orchid Research Laboratories Ltd, Chennai, India

Inflammatory bowel disease is a chronic inflammatorycondition of the gastrointestinal tract that manifests asulcerative colitis and Crohn disease1,2. We have focusedour approach towards the design and synthesis of novelsmall molecule inhibitors for treating inflammatory boweldisease.

Several analogs of I were synthesized and screened foractivity using TNF-a as a preliminary screen followed byfinding the mechanism of action (lead compound

S106 Inflamm. Res., Supplement 2 (2008)

Page 45: IRA_15th_Conference_21.-24.9.2008

OCID2287 was found to be a potent thromboxanesynthase inhibitor with nanomolar IC50).

DRC of the lead molecule was performed using DSSinduced IBD model in mice, TNBS induced IBD in ratmodel, and Oxazolone induced colitis in mice. DiseaseActivity Index (DAI) in DSS induced IBD model in miceshowed the potency of the lead compound OCID2287 at0.1mg/kg comparable to that of Sulfasalazine at 100mg/kg.

To summarize, several analogs of I were synthesized andscreened for thromboxane synthase activity followed bythe activity in IBD disease models which facilitated theidentification of a lead compound OCID2287.

References:

1. R.S.Blumberg and W. Strober, Journal of AmericanMedical Association, 2001, 285, 643-647

2. M.G. Neumann, Romanian Journal of Gastroenter-ology, 2004, Vol.13, No.4, 309-316

A146

L-17 SIGNALING INDUCES SEQUENTIALPHOSPHORYLATION OF C/EBPb

Fang Shen1,*, Nan Li2, Troy Wood2, and Sarah L. Gaffen1

1Dept. of Oral Biology, 2Dept. of Chemistry, SUNYBuffalo. 3435 Main St. , Buffalo NY 14214.

IL-17 induces target gene expression via activation of thetranscription factors NF-kB and C/EBPb. C/EBPb issubject to many posttranslational modifications. To inves-tigate phosphorylation of C/EBPb following IL-17signaling, ST-2 stromal cells were stimulated by IL-17and C/EBPb was enriched by immunoprecipitation. Afterin-gel trypsin digestion, ESI nano-spray tandem MSspectrometry was used to evaluate phosphorylationsites. Within 1 hour, IL-17 stimulation induced sequentialphosphorylation of two distinct Thr/Ser residues in theregulatory domain of C/EBPb. IL-17-induced C/EBPbphosphorylation was completely blocked by an ERKspecific inhibitor. Fibroblasts expressing the IL-17RAreceptor V553H mutation, which is defective in activatingthe MAPK pathway, also failed to induce C/EBPbphosphorylation after IL-17 stimulation. Thus, IL-17triggers multiple phosphorylation events on C/EBPbthat correlate with signaling function.

A147

MODULATORY ROLE OF2-ACETAMIDOPHENOL ON THE EXPRESSIONOF CD44 CELL SURFACE MARKERS IN THEBRAIN OF ADJUVANT INDUCED ARTHRITICMODEL (AIA) OF RATS

Durr-e-Shahwar Momin1, Sameera Khurshid1, HumaJawed1, Kiran Aslam1, S.A. Saeed2 and Shabana U.Simjee*1

1Pharmacology Unit, H.E.J. Research Institute ofChemistry, 2Dr. Panjwani Centre for Molecular Medicineand Drug Research, International Center for Chemical andBiological Sciences, University of Karachi, Karachi-75270,Pakistan. Email: [email protected], [email protected]

Rheumatoid Arthritis is a chronic inflammatory auto-immune disease involving damage to the joints andsynovial tissue. The inflammatory process is triggered byup regulation of cell surface marker CD44 inflammatorycells especially during the progression of inflammation inrheumatoid arthritis. In addition, the interaction betweenCD44 and extracellular hyaluronan has been reported tobe involved in a variety of pathological processes at thesite of inflammation. In the present study, we haveinvestigated the effect of 2-acetamidophenol (HOC6H4

NHC(O)CH3) on the expression of CD44 markers and itspotential role as anti-arthritic agent in AIA rats. To ourknowledge, much of the work done on the CD44 up-regulation in case of rheumatoid arthritis were carried outin the synovial fluid aspirate from the inflamed joint.However, we have evaluated the effect of 2-acetamido-phenol on the expression of CD44 in the brain of arthriticrats. The CD44 immunohistochemistry was done usingmonoclonal anti-CD44 antibodies. Our results demon-strate a marked decrease in the CD44 expression in the 2-acetamidophenol treated arthritic rats compared to thearthritic control group and indomethacine treatedarthritic rats. The progression of the disease was moni-tored by measuring body weight and paw volume. Thearthritic rats treated with 2-acetamidophenol not onlyinhibited the macroscopic changes such as erythema andswelling of limbs, but also exhibited significant attenu-ation of the increase in paw volume produced as a resultof arthritis induction. There was also a slight but non-significant reduction in the body weights of 2-acetamido-phenol treated arthritic rats however it was recovered inthe end. Based on these results, we suggest that 2-acetamidophenol is not only anti-arthritic but also controlthe underlying mechanism of the inflammatory processesassociated with arthritis. In addition, the determination ofCD44 in brain samples can also be a strong predictivemarker of the inflammatory processes involved in thearthritis.

Inflamm. Res., Supplement 2 (2008) S107

Page 46: IRA_15th_Conference_21.-24.9.2008

A148

PROTECTIVE EFFECT OF NON-SELECTIVEAND SELECTIVE COX-2-INHIBITORS INHYPOXIA STRESS-INDUCED BEHAVIORALAND BIOCHEMICAL ALTERATIONS

Anant Singh1*, Amritpal Singh2, Hardevinderpal Singh2

1Chandigarh College of Pharmacy, Landran (Mohali),India2Pharmacology Division, Department of PharmaceuticalSciences and Drug Research, Panjabi University, Patiala-,India.

Hypoxia stress has been reported to produce severalbehavioral, neurochemical and biochemical alterations.Cyclooxygenase (COX) enzymes are playing a vital rolein pathogenesis of several brain disorders includingAlzheimer disease, epilepsy, depression, in addition topain and inflammation. In the present work, we examinedthe role of non-selective (naproxen) and selective (rofe-coxib, valdecoxib) COX-2 inhibitors against hypoxiastress-induced behavioral alterations and oxidativedamage in mice. Mice were subjected to hypoxia stressfor a period of 4 h. Naproxen (5, 10 and 20 mg/kg, ip),rofecoxib (5, 10 and 20 mg/kg, ip) or valdecoxib (5, 10 and20 mg/kg, ip) were administered 30 min before hypoxiastress. Four-hour hypoxia stress significantly causedanxiety-like behavior, memory deficit and impairedmotor activity as well as oxidative damage (raised lipidperoxidation, nitrite activity, depletion of reduced gluta-thione and catalase activity) as compared to naiveanimals placed on sawdust (p < 0.05). Pretreatmentwith Naproxen (5, 10 and 20 mg/kg, ip), rofecoxib (5, 10and 20 mg/kg, ip) or valdecoxib (5, 10 and 20 mg/kg, ip)significantly improved locomotor activity, antianxietyeffect, memory retention (memory deficit) and attenu-ated oxidative damage (lowering of raised malondialde-hyde, nitrite activity, restoration of reduced glutathioneand catalase activity as compared to hypoxia stress group(p < 0.05). Results propose the neuroprotective andantioxidant effect of both non-selective and selectiveCOX-2 inhibitors.

A149

ROSIGLITAZONE PREVENTSHYPERHOMOCYSTEINEMIA-INDUCEDMYOCARDIAL REMODELING THROUGH MASTCELL STABILIZATION IN RATS

Hardevinder Pal Singh, Amrit Pal Singh*, Tajpreet Kaurand Anant Singh

Department of Pharmaceutical Sciences & Drug Research,Punjabi University, Patiala-147002 E-mail: [email protected]

Objectives: The present study has been designed toinvestigate the effect of rosiglitazone, a peroxisomeproliferator receptor agonist-g (PPAR-g) in hyperhomo-cysteinemia-induced myocardial hypertrophy and fibrosisin rats. Methods: Rats were administered L-methionine

(1.7mg/kg/day p.o.) for 8 weeks to produce hyperhomo-cysteinemia. Rosiglitazone (3 and 5mg/kg/day, p.o.) treat-ment was started from the first day of administration ofL-methionine and were continued for 8 weeks in rats. Thedevelopment of myocardial remodeling was assessed interms of measuring ratio of left ventricular (LV) weight tobody weight (LVW/BW), LV wall thickness (LVWT), LVprotein content (mg/g of LV) and LV collagen content(mg/g of LV). The haematoxylin-eosin staining, picro-sirius red and toluidine blue staining was done to measurecardiomyocyte diameter, collagen deposition and themast cell density in left ventricular sections. Results:The hyperhomocysteinemia significantly increased LVW/BW, LVWT, LV protein, collagen content. Histologicalstudies revealed increased cardiomyocyte diameter,extensive fibrosis and increased mast cell density.However, rosiglitazone treatments significantly attenu-ated hyperhomocysteinemia-induced pathological cardiachypertrophy and fibrosis without changing serum homo-cysteine levels in rats. Conclusion: It may be concludedthat hyperhomocysteinemia-induced myocardial remod-eling is associated with increase in density of mast cells inheart. Moreover, rosiglitazone may have attenuatedhyperhomocysteinemia-induced pathological cardiachypertrophy by preventing the degranulation andincrease in density of mast cells.

A150

EVALUATION OF NOVEL TOPICAL DRUGDELIVERY SYSTEMS OF COLCHICINE INMONO SODIUM URATE (MSU) MODEL OFGOUT IN RATS

Amrit Pal Singh, Hardevinder Pal Singh*, Anant Singhand Subheet Jain

Department of Pharmaceutical Sciences & Drug Research,Punjabi University, Patiala-147002

The gout characterized by deposition of urate crystals,agonizing pain and inflammation of joints, is a problemaffecting world�s adult population. Colchicine widely useddrug for treatment of gout is associated with numerousside effects such as nausea, diarrhea, neuro-myopathy andbone marrow suppression. The topical drug deliverysystems offer advantages such as by-passing hepaticmetabolism, minimizing side effects and avoid drugdegradation due to gastric pH. Hence, various formula-tions such as microspheres, elastic and non-elastic lipo-somes and niosomes were prepared and evaluated usingmono sodium urate model of gout in rats. The subcuta-neous air pouch was formed in anaesthetized rats byadministering 10 mL of sterile air. The sterile air was re-injected in air pouch every 2-3 days to maintain pseudo-gout conditions. After 6 days, rats were divided into MSUcontrol (without drug treatment), oral colchicine treat-ment and groups receiving colchicine entrapped inmicrospheres, niosomes, elastic and non-elastic liposomes(n=10-12/group). Rats from each group were sacrificedafter 6, 12 and 24 hours of MSU administration andevaluated for exudate volume of air pouch, total leuko-cyte count (TLC). Moreover, haematoxylin-eosin stainingwas done for gross histology to observe extent of tissue

S108 Inflamm. Res., Supplement 2 (2008)

Page 47: IRA_15th_Conference_21.-24.9.2008

damage and collagen deposition considered as marker forfibrosis was assessed using picrosirius red staining. TheMSU administration in air pouch induced extensive fluidaccumulation, increased TLC, bizarre tissue and exten-sive fibrosis. The elastic liposomes claimed best efficacyfollowed by non elastic liposomes, niosomes, micro-spheres and oral administration of colchicine. Hence,our finding suggest that novel topical drug deliverysystems especially elastic liposomes have the potentialto used for sustained and site specific delivery ofcolchicine than oral therapy.

A151

GADOLINIUM DECREASES INFLAMMATIONRELATED TO MYOCARDIAL ISCHEMIA ANDREPERFUSION INJURY

J. L. Strande*, K. V. Routhu, A. Hsu, A. C. Nicolosi, J. E.Baker.

Medical College of Wisconsin, Milwaukee, Wisconsin53226.

Gadolinium (Gd) protects the heart against infarctionfollowing ischemia and reperfusion (I/R). We determinedthe impact of Gd treatment on monocyte and neutrophilfunction during I/R. Rats (n=6/gp) were treated withsaline or Gd (20 mmol/kg) and subject to I/R. Sham ratswere not subject to I/R. I/R resulted in a 2-3 fold increasein circulating monocytes and neutrophils, and increasedmyocardial GM-CSF, IL-1, IL-8, TNF-a, and myeloper-oxidase (MPO) activity. Gd decreased the number ofcirculating monocytes and neutrophils after I/R to levelsbelow those present prior to ischemia. Gd decreased theproduction of GM-CSF and IL-1 in the myocardium butnot IL-8 and TNF-a after I/R. Furthermore, Gddecreased MPO activity after I/R to levels below thosemeasured in the Control or Sham groups. Gd treatmentprior to I/R decreases circulating monocytes and neutro-phils, macrophage secreted cytokines, and neutrophilinfiltration into injured myocardium. These resultssuggest Gd decreases leukocyte migration and activationmay be a novel treatment for inflammation duringischemia and reperfusion.

A152

THE DISCOVERY OF A SERIES OF NOVELSMALL MOLECULE MACROCYCLIC TNF-aANTAGONISTS

Nick Terrett*, Ben Benton, Timothy F. Briggs, FrankFavaloro Jr., Steve Hale, Jinbo Lee, Duke Vo, ChrisWilson, & Julian Bond.

Ensemble Discovery Corp., 99 Erie Street, Cambridge,MA 02139, USA. Email: [email protected]

We have developed an integrated platform for thesynthesis and screening of macrocyclic molecules(EnsemblinsTM) that can interact with protein-proteindrug discovery targets. Tumor necrosis factor alpha (TNF-

a) has been linked to the pathogenesis of inflammatorydisease, and agents that can prevent binding of TNF-a toits receptors have utility in the treatment of rheumatoidarthritis, Crohn�s disease, psoriasis and ankylosing spon-dylitis. Currently marketed drugs are biologicals thattarget sequestration of TNF-a, and there are very fewsmall molecule TNF-a antagonists at any stage ofdevelopment. Ensemble Discovery has recently identifieda series of selective and reversible small molecule macro-cycles that competitively antagonize the activity of TNF-aon TNF receptors in both biochemical and cell-basedassays. These compounds are currently in pre-clinicaldevelopment.

A153

IDENTIFICATION OF FUNCTIONAL ROLES FORBOTH IL-17RB AND IL-17RA IN MEDIATINGIL-25 INDUCED ACTIVITIES

Joel E. Tocker *, Erika A. Rickel, Lori A. Siegel, Bo-Rin P.Yoon, James Rottman, David Kugler, Michael R. Comeau ,and Alison L. Budelsky

*Departments of Inflammation Research and Pathology,Amgen, Seattle WA 98119

IL-25 (IL-17E) is a unique IL-17 family ligand thatpromotes Th2-skewed inflammatory responses. Intranasaladministration of IL-25 into naXve mice induces pulmo-nary inflammation similar to that seen in patients withallergic asthma, including increases in BALF eosinophils,BALF IL-5 and IL-13 concentrations, goblet cell hyper-plasia, and increased airway hyperresponsiveness. IL-25has been reported to bind and signal through IL-17RB(IL-17BR, IL-17Rh1). It has been demonstrated recentlythat IL-17A signals through a heteromeric receptorcomposed of IL-17RA and IL-17RC. We sought todetermine whether other IL-17 family ligands also utilizeheteromeric receptor complexes. The required receptorsubunits for IL-25 biological activities were investigatedin vitro and in vivo using a combination of knockout (KO)mice and antagonistic antibodies. Unlike wild-type mice,cultured splenocytes from either IL-17RB KO or IL-17RA KO mice did not produce IL-5 or IL-13 in responseto IL-25 stimulation, and both IL-17RB KO and IL-17RAKO mice did not respond to intranasal (IN) administra-tion of IL-25. Furthermore, treatment with antagonisticmonoclonal antibodies to either IL-17RB or IL-17RAcompletely blocked IL-25 induced pulmonary inflamma-tion and airway hyper-responsiveness (AHR) in naXveBALB/c mice, similar to the effects of an antagonisticantibody to IL-25. Finally, a goat blocking antibody tohuman IL-17RA prevented IL-25 activity in a primaryhuman cell based assay. These data demonstrate for thefirst time that IL-25 mediated activities require both IL-17RB and IL-17RA, and provide another example of anIL-17 family ligand that utilizes a heteromeric receptorcomplex.

Inflamm. Res., Supplement 2 (2008) S109

Page 48: IRA_15th_Conference_21.-24.9.2008

A154

THE ANTI-OBESITY EFFECT OFGYEONGSHINHAEGIHWAN T2 IS ASSOCIATEDWITH A DECREASED NITRIC OXIDESYNTHESIS

Hong-Kun Rima, Hyo-Jin Ana, Jin-Woo Hongb, Hyun-JaJeongc, Seung-Heon Hongd, Hyung-Min Kima, Jae-YoungUma,*

aCollege of Oriental Medicine, Institute of OrientalMedicine, Oriental Medical Science Center, Kyung HeeUniversity, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul,Republic of Korea, bDepartment of Cardiovascular andNeurologic Diseases (Stroke Center) College of OrientalMedicine, Kyung-Hee University, Seoul, Republic ofKorea, cBiochip Research Center, Hoseo University, 165,Sechul-ri, Baebang-myun, Asan, Chungnam, Republic ofKorea, dCollege of Pharmacy, Wonkwang University,Iksan, Jeonbuk, Republic of Korea

GyeongshinhaeGihwan T2 (GGT2) is a newly developedoriental medicine to help control weight. To evaluate theanti-obesity effect of GGT2, we investigated a possibleeffect of GGT2 on macrophage-related obesity reactions;nitric oxide production and cytokine secretion in mouseperitoneal macrophages. According to recent reports,macrophages are participated in fat accumulation andclosely related with obesity. In this study, using mouseperitoneal macrophages, we have examined whetherGGT2 affects the production of nitric oxide (NO),tumor necrosis factor-a (TNF-a), and interleukin (IL)-12 by interferon-g and lipopolysaccharide (LPS). GGT2inhibits LPS-induced NO production in a dose-dependentmanner. The decrease in NO synthesis was reflected as adecreased amount of inducible NO synthase protein. Wealso found that GGT2 inhibits pro-inflammatory cyto-kines, TNF-a and IL-12 production. In mouse embryopreadipocyte 3T3-L1, GGT2 reduced the viability in adose-dependent manner. These findings mean that GGT2can be used in preventing and controlling adipogenesisand obesity.

A155

EFFECTIVE MECHANISM OF HERB COMPLEXPRESCRIPTION ?ANSSICHEGAMSAN9 INOBESITY

Jin-Woo Hongb, Hyo-Jin Ana, Hong-Kun Rima, Hyun-JaJeongc, Seung-Heon Hongd, Hyung-Min Kima, Jae-YoungUma,*

aCollege of Oriental Medicine, Institute of OrientalMedicine, Oriental Medical Science Center, Kyung HeeUniversity, 1 Hoegi-Dong, Dongdaemun-Gu, Seoul,Republic of Korea, bDepartment of Cardiovascular andNeurologic Diseases (Stroke Center) College of OrientalMedicine, Kyung-Hee University, Seoul, Republic ofKorea, cBiochip Research Center, Hoseo University, 165,Sechul-ri, Baebang-myun, Asan, Chungnam, Republic ofKorea, dCollege of Pharmacy, Wonkwang University,Iksan, Jeonbuk, Republic of Korea

The Anssichegamsan (ACS) is a newly developed dietaryproduct to help control weight. The aim of this study wasto evaluate whether ACS combined with a high fat (HF)diet could influence body weight, fat accumulation andglucose level in blood. Rats were fed for 6 weeks withstandard diet, HF diet, and HF + 10% ACS diet. Bodyweight was recorded weekly, and plasma levels of totalcholesterol, triglyceride, and glucose were analyzed at theend of the study. Weight increases in the 10% ACS groupwere significantly less than in the HF diet group (P <0.05). Plasma triglyceride level and total cholesterol levelwere significantly decreased by 15.0%, and 24.2% in ACSdiet group. Glucose level also was decreased by 61.8% inACS group compared with HF diet group. In addition,ACS inhibited obesity related cytokines, such as inter-leukin (IL)-6, tumor necrosis factor-a, and IL-1b in rats�serum. In vitro, the author investigated ACS can preventthe differentiation of preadipocyte to adipocyte. Trigly-ceride contents of adipocytes were also inhibited by ACSsignificantly, and these effects were through inhibition ofPPARg expression. These findings indicate that ACS maybe beneficial in the reduction of HF diet-induced over-weight.

A156

EXPLORATION OF THE MAP3K TAK1 AS ATARGET FOR MODULATING INFLAMMATORYARTHRITIS

F.A.J. van de Loo*, J. Geurts, W.B. van den Berg

Rheumatology Research & Advanced Therapeutics,Nijmegen, the Netherlands

In various human fibroblasts, TGF-b-activated kinase-1(TAK1) has been demonstrated as a pivotal upstreammediator of NF-kB, p38 MAPK and JNK activationinduced by IL-1, TNFa and TLR2/4 ligands. Therapeuticapplicability of TAK1 as a target in synovial fibroblastswas tested using the small molecule inhibitor 11,12-dihydro-5Z-7-oxozeaenol (BRON). At a concentration of1 mM, a significant reduction was observed for IL-1b-induced activation of the NF-kB and C/EBPb-dependentSAA3 promoter. Using a lentiviral 5xNF-kB-luciferasereporter it was shown that this reduction was notmediated through a diminished NF-kB activation.Despite the lack of blocking NF-kB, TAK1 inhibitionproved very effective in preventing IL-1b-induced upre-gulation of matrixmetalloproteinases (MMP-1,3,13)expression and markedly reduced the upregulation ofchemokines (IL-8, MCP-1) and IL-6. Since synovialfibroblasts are the main producers of these genes in theinflamed joint, targeting TAK1 using small moleculeinhibitors or gene therapy overexpressing a dominant-negative mutant of TAK1 hold promising potential intreatment of inflammatory arthritis.

S110 Inflamm. Res., Supplement 2 (2008)

Page 49: IRA_15th_Conference_21.-24.9.2008

A157

TREATMENT WITH ANTI-CD30 LIGANDPREVENTS DISEASE PROGRESSION INMURINE SYSTEMIC LUPUS ERYTHEMATOSUS

Cynthia R. Willis*, Yi-Ling Hu, Anh Leith, and James B.Rottman

Departments of Inflammation and Pathology, Amgen Inc.USA

The cell-surface glycoproteins CD30 and CD30 ligand(CD30L) are expressed primarily on activated immunecells, and their interactions regulate T cell responses.Systemic lupus erythematosus (SLE) is an autoimmunerheumatic disease exhibiting many immunologic abnor-malities. Similar to human SLE, disease expression inNZB/W F1 mice requires activated T cells to stimulate Bcells that secrete high affinity IgG autoantibodies. Weused flow cytometry to determine the kinetics and cellularexpression of CD30L during the course of lupus disease inuntreated NZB/W F1 mice, and we compared the abilityof CD30L-blocking/depleting and CD30L-blocking anti-bodies to prevent lupus disease progression in these mice.CD30L was expressed on subpopulations of leukocytesduring the course of disease. The progression of lupusdisease was prevented by treatment with CD30L anti-bodies. However, a CD30L blocking antibody withdepleting properties was superior to simply blockingCD30/CD30L interactions. These data suggest that deple-tion of CD30L+ cells may be a viable treatment forhuman SLE patients.

A158

ELIMINATION OF TOLMETIN ULCERS BYANISODAMINE (ANSA)

S. Wong* & J. P. Devlin

Healing Care, Ltd, Lansdale, PA, 19446

The efficacy of Tolmetin and other NSAIDs have beenshown to be enhanced by acetaminophen (APAP).However APAP has no significant effect on the ulcer-ogenicity of NSAIDs (Wong & Gardocki, 1983). AllNSAIDs ,including Selective COX-2 Inhibitors, haveulcerogenic activity, leading the Food and Drug Admin-istration (FDA) to withdraw Vioxx from the market andmandated warning labels added to all NSAID packageinserts. Therefore it has become extremely important tofind a non-ulcerogenic AI agent.Recent investigations by Healing Care, Ltd., discoveredthat the ulcerogenicity of NSAIDs were suppressed byTropane alkaloids, atropane, scopolamine, and anisod-amine (ANSA).

Careful studies, using a five-day rat ulcerogenicity assay,showed that Tolmetin ulcers could be eliminated byANSA. The ID50 and ID100 values (53.7 and 148 mg/kg/day, p.o. respectively) were determined. Further studieswith Ibuprofen, Naproxen and Piroxicam generatedsimilar activity profiles.

Conclusion: “ Ulcer induction by NSAIDs can beantagonized or eliminated by ANSA, a Tropane alka-loid”.

A159

SMALL MOLECULE CXCR2 ANTAGONISTPREVENTS HYPEROXIA-INDUCEDNEUTROPHIL ACCUMULATION IN THE LUNGSOF NEWBORN RATS

E. Yurkow*1, M. Winters1, C. Crysler1, N. Subasinghe1, D.Ryan1, L. Leong1, S. Zhao1, R. Donatelli1, M. Mazzulla1,L. Boczon1, C. Manthey1, C. Molloy1, H. Raymond2, L.Murray2, L. McAlonan2 & B. Tomczuk1

1Johnson & Johnson Pharmaceutical Research &Development, Spring House, PA; 2Centocor Research &Development, Radnor, PA

Recruitment of neutrophils is a normal physiologicalresponse to infection and tissue damage. However,excessive numbers of these cells can exacerbate tissuedamage by releasing proteases, oxygen radicals, and othermediators contributing to the development and severityof conditions such as COPD and acute respiratory distresssyndrome. CXC chemokines interact with CXCR1 &CXCR2 and are known to mediate, in part, the recruit-ment of neutrophils to areas of lung injury. A series ofacylsulfamide derivatives were identified that function ascompetitive, reversible small molecule inhibitorstargeting CXCR2. A lead compound in the seriesexhibited an IC50 of 50nM against Eu-IL-8/CXCR2membrane binding, inhibited IL-8-induced calcium fluxin cells expressing hCXCR2 (IC50 = 5nM) and inhibitedrabbit neutrophil GROalpha-driven chemotaxis. Thiscompound also demonstrated human liver microsomalstability, low clearance (3.8 mL/min/kg with t1/2 = 2.7h)and excellent oral bioavailability (107%). In vivo, thecompound was active at preventing neutrophil accumu-lation in the lungs of newborn rats exposed to hyperoxicconditions. These results support the continued evalua-tion of CXCR2 antagonists as therapeutic agents indiseases where neutrophil–mediated exacerbation ispresent.

A160

SYNERGISTIC INDUCTION OF IL-10 BY A TLRAGONIST AND A PHOSPHO-CERAMIDEANALOG IS MEDIATED BY CAMP

Dorit Avni, Meir Goldsmith, and Tsaffrir Zor*

Department of Biochemistry, Tel-Aviv University,Tel-Aviv, Israel.

Expression of the anti-inflammatory cytokine interleukin10 (IL-10) can be induced either by toll-like receptor(TLR) agonists such as LPS, or by various endogenousstimuli, in particular those acting via a cAMP-dependentsignaling pathway. Phospho-ceramide analog-1 (PCERA-1): 1-methyl-2-(3-methoxyphenyl)-2-(octanoylamino)ethyl-

Inflamm. Res., Supplement 2 (2008) S111

Page 50: IRA_15th_Conference_21.-24.9.2008

disodium-phosphate, down-regulates LPS-induced produc-tion of TNFa in macrophages in a cAMP-dependentmanner. The objective of this study was to evaluate theeffect of PCERA-1 on IL-10 production, and to determineits mechanism. We show here that PCERA-1 induces IL-10production in synergism with various TLR agonists.Cooperativity is evident both at the mRNA and proteinlevels. IL-10 production by LPS and/or PCERA-1 ismediated by PKA and the activity of PCERA-1 can bemimicked by a cell-permeable analog of cAMP. Further-more, in the absence of PCERA-1, the residual IL-10induction by LPS is completely blocked by the b-adrenergicreceptor antagonist, propranolol. Our results thus indicatethat basal cAMP is essential for IL-10 induction by LPSand that a co-stimulus by a TLR agonist and a cAMP-elevating agent results in synergistic IL-10 production.

S112 Inflamm. Res., Supplement 2 (2008)

Page 51: IRA_15th_Conference_21.-24.9.2008

Van Arman Award Competition Abstracts

The Inflammation Research Association sponsors acompetition for the encouragement of young scientiststo perform exploratory and applied research in thegeneral area of inflammation. Contestants must becandidates for advanced degrees: M.S., Ph.D., M.D.,D.O., D.D.S., D.V.M., etc., or first year post-doctoralfellows. Those who have won first place in a previous yearare ineligible to compete again.These awards are in recognition of the late C. GordonVan Arman, who had a long and distinguished career asan industrial scientist, during which he published over 100scientific papers. The development of the drugs diphe-noxylate, disopyramide, sulindac, and diflunisal can be

directly attributed to his work. In 1970, Dr. Van Armanwith Edward Takesue, Marvin Rosenthale, and Mary LeeGraeme founded the Inflammation Research Associationas an informal forum for bench scientists to exchangeresearch ideas in inflammatory diseases. Through thisaward, the IRA wishes to develop a commitment to highquality inflammation research in young scientists.Prior to the Conference, the Scholarship Committeeselects the five finalists based on submitted min-papers.Finalists will attend the Conference and participate inposter and oral presentations to the committee. Based onthese presentations and the mini-papers, awards will bepresented to the finalists.

Page 52: IRA_15th_Conference_21.-24.9.2008

VA01

DIFFERENTIAL MODULATORY EFFECTS OFTLR2 AND TLR4 ON T CELL BALANCE INEXPERIMENTAL ARTHRITIS: POSSIBILITIESFOR NEW THERAPEUTIC STRATEGIES

Shahla Abdollahi-Roodsaz*, Leo Joosten, Fons van deLoo and Wim B. van den Berg

Rheumatology Research and Advanced Therapeutics,Radboud University Medical Centre Nijmegen, TheNetherlands;e-mail : [email protected]

Toll-like receptors (TLRs) might contribute to theprogression of rheumatoid arthritis through recognitionof microbial or host-derived ligands found in arthriticjoints. We investigated the involvement of TLR2 andTLR4 in the progression of arthritis using IL-1 receptorantagonist-knockout (IL1Ra-/-) mice, which spontane-ously develop an autoimmune T cell-mediated arthritis.Clinical and histopathological evaluation of IL1Ra-/-

TLR2-/- mice revealed more severe arthritis, characterizedby reduced suppressive function of regulatory T cells(Tregs) and substantially increased IFN-gamma produc-tion by T cells. IL1Ra-/-TLR4-/- mice were, in contrast,protected against severe arthritis and had markedly lowernumbers of Th17 cells and a reduced capacity to produceIL-23/IL-17. Therapeutic treatment of both IL-1Ra-/- andcollagen-induced arthritis models using a TLR4 antago-nist prevented joint inflammation and cartilage and bonedestruction.

These studies demonstrate distinct roles of TLR2 andTLR4 in the regulation of inflammatory cytokines and Tcell balance, and indicate that TLR4 might be a potentialtherapeutic target in the treatment of RA.

VA02

THE ROLE OF CC CHEMOKINE RECEPTOR 7DURING INVASIVE ASPERGILLOSIS

Adam Hartigan*, Cory Hogaboam

University of Michigan Medical School, Department ofPathology, Ann Arbor, Michigan

Aspergillus fumigatus is ubiquitously found in the envi-ronment and is easily cleared from the body in immuno-competent hosts. Invasive aspergillosis (IA) develops inimmunocompromised patients, particularly in transplantrecipients, and is a leading cause of death in patients whohave undergone allogeneic hematopoietic stem cell trans-plantation (HSCT). Chemokine receptor 7 (CCR7) andits ligands, CCL19 and CCL21, are responsible for themigration of dendritic cells (DCs) from sites of infectionand inflammation to secondary lymphoid organs. Thus, ithas been suggested that CCR7 plays an important role indevelopment of the adaptive immune response, whilequestions remain regarding the role of CCR7 during aprimary immune response. We are currently investigatingthe role of CCR7 expression on DCs during an IA

infection using a murine HSCT model. Mice receivingCCR7 deficient stem cells have increased survival anddecreased lung pathology compared to mice transplantedwith wild type stem cells. Flow cytometric analysis of micereceiving wild type or CCR7 deficient stem cells, revealsan increase in the number of DC present in the lungs ofthe knockout animals following infection with aspergillusconidia. Additionally, RNA data shows an increase inIFN-g, TNF-a, IP-10, and IDO in the mice receivingCCR7-/- cells. Our results suggest that the absence ofCCR7 provides protection from IA after myeloablationand stem cell reconstitution. This data revels a potentialrole for CCR7 in a DC mediated primary immuneresponse against aspergillus fumigatus during IA.

VA03

PRO-INFLAMMATORY CYTOKINES INHIBITCHONDROGENESIS OF HUMANMESENCHYMAL STEM CELLS THROUGH NF-kBDEPENDENT PATHWAYS

Ryan M. Porter*, Nathalie Wehling, Glyn D. Palmer, andChristopher H. Evans

Center for Molecular Orthopaedics, Harvard MedicalSchool, Boston, MA

The differentiation of mesenchymal stem cells (MSCs)into chondrocytes provides an attractive basis for theregeneration of articular cartilage, but chondrogenesiswill often need to occur in the presence of inflammatorymediators produced in response to injury or disease. Herewe examined the effect of two important inflammatorycytokines, interleukin-1b (IL-1b) and tumor necrosisfactor-a (TNF-a), on the chondrogenic behavior ofhuman MSCs. Aggregate cultures of MSCs recoveredfrom the femoral intermedullary canal were used. Chon-drogenesis was evaluated by measuring proteoglycan andcollagen synthesis at both the protein and message levels.The possible involvement of NF-kB in mediating IL-1beffects was assessed by adenoviral delivery of a dominantnegative inhibitor of NF-kB (srIkB). Both IL-1b andTNF-a inhibited hMSC chondrogenesis in a dose-dependent manner, which was associated with a markedactivation of NF-kB. Delivery of srIkB abrogated theactivation of NF-kB and rescued the chondrogenicresponse. Strategies for enabling cell-based cartilagerepair within inflammed joints include targeting notonly individual pyrogens, such as IL-1 and TNF, butalso important intracellular mediators, such as NF-kB.

S114 Inflamm.Res., Supplement 2 (2008)

Page 53: IRA_15th_Conference_21.-24.9.2008

VA04

BZ-423 IMPROVES GVHD-ASSOCIATED TISSUEDAMAGE AND MORTALITY BY SPECIFICALLYTARGETING EFFECTOR T CELLS

Daniel R. Wahl*, Gary D. Glick, and James L.M. Ferrara

University of Michigan, Ann Arbor, Michigan 48109

Graft-versus host disease (GVHD) is the major compli-cation of allogeneic bone marrow transplantation andcauses systemic inflammation, which can be fatal if leftuntreated. The treatment of GVHD with High-dosecorticosteroids results in complete responses in less thanhalf of patients and often leads to severe immunosup-pression and other serious consequences. We have inves-tigated Bz-423, a novel therapeutic agent that targets themitochondrial ATPase, in a model of acute GVHD. Wefound that Bz-423 treatment beginning on day 3 aftertransplant prevented GVHD-associated mortality andreduced donor T cell infiltration into the liver and bonemarrow, two GVHD target organs. Importantly, Bz-423did not impair donor T cell expansion or activationmarker expression, indicating that, unlike High-dosecorticosteroids, Bz-423 is not broadly immunosuppressive.

VA05

MAPPING THE BINDING OF MACROPHAGEMIGRATION INHIBITORY FACTOR (MIF) TOTHE CHEMOKINE RECEPTOR CXCR4

Swen Zierow*, James Murphy, Melanie Merk, Micheal E.Hodsdon, and Elias Lolis

Yale University School of Medicine, New Haven,Connecticut, 06520

The cytokine macrophage migration inhibitory factor(MIF) plays a critical role in many acute and inflamma-tory diseases. MIF acts as a major regulator of inflamma-tory cell recruitment and atherogenesis. These chemo-kine-like functions of MIF were recently shown to bemediated through interaction to the chemokine receptorsCXCR4 and CXCR2. However, the molecular details ofthis interaction have not yet been determined. Wedescribe herein a peptide derived from the N-terminalextracellular region of the CXCR4 receptor as a site ofinteraction with human MIF and Leishmania major MIF.From 1H15N chemical shift perturbation studies a directbinding interaction between MIF and the N-terminal 27residues of CXCR4 is shown. Titration studies usingsteady-state fluorescence spectroscopy result in a disso-ciation constant of 3.1�0.8 mM. Notably, MIF-triggeredmonocyte chemotaxis activity is ablated by this N-terminal CXCR4 peptide. Structural studies of the MIF/CXCR4 complex may aid in the design of new drugstargeting MIF related diseases.

Inflamm.Res., Supplement 2 (2008) S115

Page 54: IRA_15th_Conference_21.-24.9.2008
Page 55: IRA_15th_Conference_21.-24.9.2008

Author Index(* denotes presenting author)

Abdollahi-Roodsaz*,S. VA01Abdollahi-Roodsaz, S. A122Abdollahi-Roodsaz, S. A123Abreu*, M.T. SA04Adler*, K.B. SA24Aggarwal, R. A116Ahrens, E.T. A112Akama, T. A139Albers, R. A140Ali, I. A101Alley, M.R.K. A139Alt*, C. A100Amalfitano, A. A138Amir*, M. A101An, H-J. A119An, H-J. A120An, H-J. A154An, H-J. A155Anjum, S. A118Antunes, J. A139Appledorn, D.M. A138Aran, J.M. A135Arnett, H.A. SA01Aslam, K. A147Audoly, L. SA13Audoly. L. A109Avni, D. A160Awan, S.I. A118

BaileyHealy, I. A103Baillie*, R.A. A102Baillie, R. A116Baker, J.E. A151Banfield, C. SA22Baugh, J. A127Bekkali, Y. A125Bendele, A. A134Bennett, B. A140Benton, B. A152Bentzien, J. A125Berg, E.L. A132Berson*, A.E. A103Bhagwat, S. A140Bhakiaraj, D. A145Bilter, G. A140Bishop, G.A. A128Boczon, L. A159Bolognese, B.J. A136Bond, J. A152Bose*, S. A104Brameld, K. A106Brenner, D. A140

Briggs, T.F. A152Brombacher, F. A113Brovarney, M. A103Brown, S.A. A106Bryant*, S. A105Bryant, S. A114Bucala, R. A127Budelsky, A.L. A153Buhr, C. A140Burgess*, L.E. A106Burnes, L.A. SA11Burnet, M. A121Burnet, M. A142Burnet, M. A143Burnette*, B. A107

Calderwood, D. A114Cao, Y. A117Capper-Spudich, E.A. A136Castaneda, J. A109Castellino*, F. SA14Chagnovich*, D. A108Chantry, D. A106Chao, Q. A140Chen*, B. A109Chen*, G.G. A110Chen, C. A111Chun, S.L. A110Cibotti, R. A109Collins*, D. A111Comeau, M.R. A153Cornicelli*, J. A112Cory Hogaboam VA02C=t>, B. SA13Cox*, G. SA23Coyle, A.J. A109Crandall, T. A141Crysler, C. A159Cutler, A. A113

D?Andrea, A. A100D?Sidocky, N. A140Dabbagh, K. A103De Groot*, A.S. SA15Denning, T.L. SA02Devesa, I. A122Devesa, I. A123Devlin, J.P. A158Donatelli, R. A159Ducharme, Y. SA13Dugo, L. A143Dyer, R. D. A108

Ehlers*, S. A113Evans, C.H. VA03

Fasciano, S. A126Favaloro Jr., F. A152Feeney, E. SA08Fellows, J. A108Ferrara, J.L.M. VA04Ferri, R. A140Foley, J.P. A136Freund, Y. A139Friedman, G. A140Friesen, R.W. SA13Fuentes, M.E. A103

Gaffen*, S.L. SA18Gaffen, S.L. A146Gan, L. A126Garssen, J. A115Getz*, G.S. SA17Geurts, J. A156Glick, G.D. VA04Glodek, A. A109Goldsmith, M. A160Goudreau, R. A114Grimshaw, C. A140Groneberg, R.D. A106Guse, J-H. A121

Hale, S. A152Han, Y. SA13Hart*, M. A114Hart, M. A105Hartigan*, A. VA02Hartog*, A. A115Havekes, L. A129Heitmann, L. A113Herbst, R. A109Ho*, R. A116Hoag, K.A. A138Hodsdon, M.E. VA05Holan, V. A143Hçlscher, C. A113Hong, J-W. A119Hong, J-W. A120Hong, J-W. A154Hong, J-W. A155Hong, S-H. A119Hong, S-H. A120Hong, S-H. A154Hong, S-H. A155Horner, M. SA22

Page 56: IRA_15th_Conference_21.-24.9.2008

Horrigan, S.K. A109Horstmann, R.D. A113Hsu, A. A151Hu, Y-L. A157Huang*, Y. A117Hunt, K.W. A106Hussain, Z. A144Hyland, D. A105Hyland, D. A114

Iacomini*, J. SA07Ikeuchi, M. SA11

Jain, S. A150Jawed*, H. A118Jawed, H. A144Jawed, H. A147Jeong, H-J. A119Jeong, H-J. A120Jeong, H-J. A154Jeong, H-J. A155Ji, L.L. A117Johnson, E. A143Johnson, Y. A110Jones, R. A142Joosten, L. VA01Joosten, L.A.B. A122Joosten, L.A.B. A123Jungbluth, G. A107

Kaddurah-Daouk, R. A102Kahn, J. A125Kaimal, V. A112Karaoglu Hanzatian, D. A104Kaur, T. A149Kavathas, P. A127Kaymakcalan, Z. A104Kehlet, H. A133Khatchenko, O. A140Khurshid, S. A147Kiener, P.A. A109Kim*, H-M. A119Kim*, H-M. A120Kim, H-M. A154Kim, H-M. A155Kim, K-Y. A120Kimura, R. A139Koch*, P. A121Koch, K. A106Koch, K. A134Koenders*, M.I. A122Koenders*, M.I. A123Kohm, A. SA08Kois, A. A140Kolker, S.J. SA11Kou, J.P. A136Krupinski, J. A135Kugler, D. A153Kulkulka, A. J. A125Kunkel, E.J. A132Kurumbail, R. A107

Lagerweij, T. A129Lagerweij, T. A131Laufer, S. A143

Laufera, S. A121Laurence*, A. SA21Le*, Y. A124LeFnez, S. A137Lee*, T.W. A125Lee, J. A152Lee, P. A106Lee, P. A134Lee, S.J. A127Lees, M. A142Leimgruber, R.M. A107Leisten, J. A140Leith, A. A157Leong, L. A159Lew, W. A140Lewis, A. A140Li*, L. A126Li, N. A146Lin*, S-L. SA22Lin, M. A141Liu, D. A109Lolis, E. VA05Long, M. W. A108Lopez, L. A134Lu*, J. SA16Lubberts*, E. SA20Luo, X. SA08

Maes, D. A111Mailliard, R.B. A112Maitra, U. A126Mancini, J. A142Mancini, J. SA13Manning*, T. SA26Manning, A. A140Mansfield, L.S. A138Manthey, C. A159Mao, C.P. A103Mao, S.H. A117Maples, K. A139Mareska, D.A. A106Marijnissen, R.J. A122Marijnissen, R.J. A123Martin, A. SA08Mathieu, S. A105Mazzulla, M. A159McAlonan, L. A159McCarrick, M. A140McCarthy, D. SA08McConville, P. A112McEvoy, J. A102McKenzie, A.N.J. A113McQueney, M.S. A136Melrose, J. A132Merk*, M. A127Merk, M. VA05Meyer, C.G. A113Miller*, S. D. SA08Mnich, S. A107Mogil*, J.S. SA10Molloy, C. A159Momin, D. A147Monahan, J. A107Monterosso, T. A112Moradi, V. A143

Morris, C. A109Motiwala, A. A140Muir, J. A140Muizzuddin, N. A111Munroe*, M.E. A128Murphy, J. VA05Murray, L. A159Murtaza, A. A105Murtaza, A. A114

Nabozny, G. A125Nadolny, L. A140Nagelkerken*, L. A129Nagelkerken*, L. A130Nagelkerken*, L. A131Naiman, B. A109Nair, R. SA02Nelson, A.D. A112Nguyen, D. A132Nicolosi, A.C. A151Nielsen, H.J. A133

O?Leary, E. A140O?Mahony*, A. A132Olivar, R. A135Olson, L. A105Olson, L. A114Omholt, P. A140Oranje, A.P. A129

Pai, S. A140Pal Singh, A. A150Pal Singh, H. A149Palmer, G.D. VA03Pavan, S. A131Pedersen, J.L. A133Persoon-Deen, C. A129Persoon-Deen, C. A130Persoon-Deen, C. A131Petersen*, L.J. A133Pheneger*, J. A134Pheneger, J. A106Picarella, D. A141Plantevin, V. A140Plattner, J. A139Plomp, A. A130Pluvinet*, R. A135Podolin*, P.L. A136Pol*, O. A137Polgar, W. A100Porter*, R.M. VA03Poy, N. A103Prasad, S. SA08Privat, S. A132Pulendran*, B. SA02

Rajagopal, S. A145Ramachandran, U. A145Rathinam*, V.A.K. A138Ravindran, P. A103Raymon, H. A140Raymond, H. A159Raza Shah, M. A144Regan, J. A125Rickel, E.A. A153

S118 Inflamm.Res., Supplement 2 (2008)

Page 57: IRA_15th_Conference_21.-24.9.2008

Riendeau, D. SA13Rim, H-K. A119Rim, H-K. A120Rim, H-K. A154Rim, H-K. A155Rock, F. A139Roitt, I. A142Rosler, E. A132Rottman, J. A153Rottman, J.B. A157Routhu, K.V. A151Ruan, L. A124Ryan, D. A159

Saeed, S.A. A147Sahasrabudhe, K. A140Saifullah, M.K. A101Sakata, S. A140Salazar, R.M. SA02Sanders*, V. A139Sanjay, K.V. A145Sapienza, J. A140Satoh,* Y. A140Savinainen*, A. A141Schlachter, S.T. A106Schopf, L. A105Schopf, L. A114Schreiber, T. A113Seed*, M. A142Seed*, M. A143Selness, S. A107Shah*, S.U. A144Sharma*, G.V.R. A145Shen,* F. A146Shevach*, E.M. SA06Shevlin, G. A140Shew, K. A100Shirley, M.A. A140Siegel, L.A. A153Simjee*, S.U. A147Simjee, S.U. A118Simjee, S.U. A144Sims, G.P. A109Singh*, A. A148Singh*, A.P. A149Singh*, H.P. A150Singh, A. A148Singh, A. A149

Singh, A. A150Singh, H. A148Skov, P.S. A133Sluka*, K.A. SA11Smith, C. SA08Smits, M. A130Soppet, D. A109Souza, D. A125Stolaki, M. A131Strande*, J.L. A151Subasinghe, N. A159Sukunath, N. A145Sumoy, L. A135Swanson, R.M. SA01

Targan*, S. SA05Terrett*, N. A152Therien, A. SA13Thirunavakkarasu, S. A145Thomson, D. S. A125Thye, T. A113Tielen, F. A131Tocker *, J.E. A153Tocker, J. SA22Toll, L. A100Tomczuk, B. A159Tong, M.C.F. A110Torres, I. A137Tran, T.T. A100Turley, D.M. SA08

Uehara, T. A140Um*, J-Y. A154Um*, J-Y. A155Um, J-Y. A120Um, J-Y. A119

van de Loo*, F.A.J. A156van de Loo, F. VA01van den Berg, W.B. A156van den Berg, W.B. A122van den Berg, W.B. A123van den Berg, W.B. VA01van der Mark. K. A130van Hasselt, C.A. A110Veldman*, T. SA25Verbeek, R. A129Verzaal, P. A129

Verzaal, P. A130Vincent, M. SA22Viney*, J.L. SA01Vlantis, A.C. A110Vo, D A152

Waegell, W. A114Wahl*, D.R. VA04Walder, R.Y. SA11Wang, D. A126Wang, J. M. A124Weaver*, C.T. SA03Wehling, N. VA03Westwick, J. A140White*, F.A. SA12Wilder*, R.L. SA09Willis*, C.R. A157Wilson, C. A152Winkler, J. A134Winters, M. A159Wixted, W.E. A136Wong*, S. A158Wood, T A146Worms, N. A130Worms, N. A131Wright*, J. SA19Wright, D. A134Wright, J. A140Wu, C. A104Wu, L. A141

Xu*, D. SA13Xu, L. A140Xu, W. A140Yi, C. A117

Yoon, B-R.P. A153Yurkow*, E. A159

Zaman, M.S. A101Zhao, S. A159Zhou, Y. A139Zierow*, S. VA05Zierow, S. A127Zong, Q. A109Zor*, T. A160Zuvela-Jelaska, L. A125

Inflamm.Res., Supplement 2 (2008) S119