University of Pennsylvania ScholarlyCommons

Publicly Accessible Penn Dissertations

Fall 2010

Developing Novel -Based Targeting Strategies for Thrombolytics

Rudy E. Fuentes The University of Pennsylvania, [email protected]

Follow this and additional works at: https://repository.upenn.edu/edissertations

Part of the Therapeutics Commons

Recommended Citation Fuentes, Rudy E., "Developing Novel Platelet-Based Targeting Strategies for Thrombolytics" (2010). Publicly Accessible Penn Dissertations. 263. https://repository.upenn.edu/edissertations/263

This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/263 For more information, please contact [email protected]. Developing Novel Platelet-Based Targeting Strategies for Thrombolytics

Abstract Use of plasminogen activators (PAs) as thrombolytic drugs is restricted to life threatening thrombotic settings because these therapies are associated with a high risk of bleeding. We hypothesize that platelet-delivered PAs would preferentially lyse nascent, pathological clots that are actively recruiting , while sparing pre-formed hemostatic clots. Two potential approaches were pursued: 1) PA- loaded platelets that release the thrombolytic from its granular stores upon activation, and 2) a thrombolytic chimeric protein that specifically binds ot human platelets and activated when the platelets are incorporated into a growing thrombus. In our first approach, we desired to develop a strategy for producing platelets ex-vivo from cultured that ectopically expressed urokinase-PA (uPA). No group had successfully produced sufficient ex-vivo generated platelets before, to side step this issue we infused ex-vivo generated megakaryocytes and showed that we can achieve a significant number of donor-derived platelets from these infused megakaryocytes in a murine model. The resulting platelets were normal in size, surface markers, circulating half-life, and were functional. Infused megakaryocytes localized to the pulmonary vasculature to shed platelets. We demonstrated, beginning with megakaryocytes derived from a transgenic mouse that ectopically express and store uPA in their alpha- granules that we can interfere with thrombosis by platelets generated from these megakaryocytes. In the second approach we produced a chimeric protein by fusing a single chain variable fragment (scFv) directed to the human-αIIb (hαIIb) platelet receptor subunit, with a human thrombin activatable pro- urokinase (uPA-T). The fusion protein (anti-PLT scFv/uPA-T) bound specifically ot human and to transgenic mice platelets that expressed hαIIb, termed hαIIb+ mice, but did not bind to wildtype (WT) mouse platelets. Anti-PLT scFv/uPA-T retained its zymogenic properties until activated by thrombin. HαIIb+ mice were protected from forming occlusive thrombi for at least 10 hrs post anti-PLT/uPA-T treatment in contrast to the short functional half-life of soluble uPA-T. Thus this dissertation presents two distinct strategies that in proof of principle studies are each promising as approaches for effective and targeted platelet directed thrombolytic, which merit further study to test clinical applicability.

Degree Type Dissertation

Degree Name Doctor of Philosophy (PhD)

Graduate Group Pharmacology

First Advisor Dr. Mortimer Poncz

Keywords Targeting thrombolytics, platelet, platelet transfusions, plasminogen activators, urokinase

Subject Categories Therapeutics

This dissertation is available at ScholarlyCommons: https://repository.upenn.edu/edissertations/263 DEVELOPINGNOVELPLATELETBASEDTARGETINGSTRATEGIES

FORTHROMBOLYTICS

RudyE.Fuentes ADISSERTATION in Pharmacology PresentedtotheFacultiesoftheUniversityofPennsylvaniainPartial FulfillmentoftheRequirementsfortheDegreeofDoctorofPhilosophy 2010 Dr.MortimerPoncz,M.D. SupervisorofDissertation VladimirR.Muzykantov,M.D.,Ph.D. GraduateGroupChairperson DissertationCommittee RodneyM.Camire,Ph.D. VladimirR.Muzykantov,M.D.,Ph.D. DougCines,M.D. DonaldL.Siegel,M.D.,Ph.D. JoelS.Bennett,M.D.

DEVELOPINGNOVELPLATELETBASEDTARGETING

STRATEGIESFORTHROMBOLYTICS

COPYRIGHT

2010

RudyE.Fuentes

ii

Acknowledgments

Iconsidermyselffortunatetohavebeeninfluencedbyanumberofbrilliantpeoplewhile pursuing my scientific career. The first person is Dr. George B. Stefano at the State

UniversityofNewYork(SUNY)OldWestbury,whogavemeanopportunitytoworkin his laboratory during my undergraduate studies, and as a consequence sparked my interest in scientific research. His mentorship provided me with a strong academic foundation that resulted with acceptance in the prestigious Pharmacology graduate programattheUniversityofPennsylvania.

During my graduate studies the most influential person responsible with the workpresentedinthisthesisismyadvisor,Dr.MortimerPoncz.Iamgratefulforgiving me the opportunity to work in his laboratory, and providing me with the invaluable guidance to analyze and take action on scientific matters. I am amazed at his vast knowledgeinhematology,andhiswayofcommunicatinganddirectinghisstudentsin thelaboratory.Iwanttoalsorecognizemycommitteememberswhohavehelpedme greatly to advance my work with their scientific insight, Drs. Rodney M. Camire ,

VladimirR.Muzykantov,DougCines,DonaldL.Siegel,andJoelS.Bennett.Another person that deserves my gratitude is Dr. Sriram Krishnaswamy, who has been instrumental in providing me with thoughtful assessments and suggestions during journalclub.

WhenIlookbackatmyacademiclife,Icansaythatithasbeenaroughride,but throughoutmyhardshipstherehasbeenmywifeKarenFuentes,whohassupportedme andencouragemetobepersistentincompletingmydegree.Iwouldalsoliketothank herforprovidingmewiththebestgiftinlife,mysonRudyA.Fuentes,andasecondone

“babyBenjamin”tobeborninthefirstweeksofFebruary2011.

iii

FinallyIwasborninthecityofLaUnioninElSalvadorandresidedinthelittle villageofElJicaroforthefirsttenyearsofmylife,untilmymotherVirginiaFuentes broughtallsixofus(Mirna,Rosa,Nelson,AnaRuth,andYolanda)totheUnitedStates

asaconsequenceofacivilwar.IamproudtosaythatIwillbethefirstpersonwitha

Ph.D. in my family, with some of my siblings achieving academic success as well. I

attribute our success to my mother’s determination in providing a better life for our

family in this country. Her perseverance has been a strong source of inspiration

throughoutmylife,andIthankherforbeingsuchaninfluentialpersontoallofus.

iv

ABSTRACT

DEVELOPINGNOVELPLATELETBASEDTARGETINGSTRATEGIESFOR

THROMBOLYTICS

RudyE.Fuentes

MortimerPoncz

Use of plasminogen activators (PAs) as thrombolytic drugs is restricted to life threateningthromboticsettingsbecausethesetherapiesareassociatedwithahighriskof bleeding.WehypothesizethatplateletdeliveredPAswouldpreferentiallylysenascent, pathological clots that are actively recruiting platelets, while sparing preformed hemostatic clots. Two potential approaches were pursued:1) PAloaded platelets that releasethethrombolyticfromitsgranularstoresuponactivation,and2)athrombolytic chimeric protein that specifically binds to human platelets and activated when the plateletsareincorporatedintoagrowingthrombus.Inourfirstapproach,wedesiredto develop a strategy for producing platelets exvivo from cultured megakaryocytes that ectopicallyexpressedurokinasePA(uPA).Nogrouphadsuccessfullyproducedsufficient exvivogeneratedplateletsbefore,tosidestepthisissueweinfusedexvivogenerated megakaryocytesandshowedthatwecanachieveasignificantnumberofdonorderived plateletsfromtheseinfusedmegakaryocytesinamurinemodel.Theresultingplatelets werenormalinsize,surfacemarkers,circulatinghalflife,andwerefunctional.Infused megakaryocytes localized to the pulmonary vasculature to shed platelets. We demonstrated, beginning with megakaryocytes derived from a transgenic mouse that ectopically express and store uPA in their alphagranules that we can interfere with thrombosisbyplateletsgeneratedfromthesemegakaryocytes.Inthesecondapproach

v

we produced a chimeric protein by fusing a single chain variable fragment (scFv)

directed to the humanαIIb (h αIIb) platelet receptor subunit, with a human thrombin activatable prourokinase (uPAT). The fusion protein (antiPLT scFv/uPAT) bound specifically to human and to transgenic mice platelets that expressed h αIIb, termed hαIIb +mice,butdidnotbindtowildtype(WT)mouseplatelets.AntiPLTscFv/uPAT retained its zymogenic properties until activated by thrombin. H αIIb + mice were protected from forming occlusive thrombi for at least 10 hrs post antiPLT/uPAT treatment in contrast to the short functional halflife of soluble uPAT. Thus this dissertation presents two distinct strategies that in proof of principle studies are each promisingasapproachesforeffectiveandtargetedplateletdirectedthrombolytic,which meritfurtherstudytotestclinicalapplicability.

vi

TableofContents

Titlepage.......................................................... i

CopyrightNotice....................................................ii

Acknowledgment....................................................iii

Abstract............................................................v

TableofContents....................................................vii

ListofTables........................................................x

ListofIllustrations...................................................xi

1. Introduction

1.1 Background

1.1.1 Pathogenesisofthrombosis..............................1

1.1.2 Historyofplateletbiology................................2

1.1.3 development..............................2

1.1.4 Plateletdevelopment....................................4

1.1.5 Plateletsroleinthrombusformation.......................6

1.1.6 Fibrinolysis............................................9

1.1.7 Keymediatorsoffibrinolysis.............................9

1.1.8 ClinicaluseofPAsandtheirlimitations....................12

1.2 Dissertationobjective.........................................14

2. Infusingmaturemegakaryocytesintomiceyieldsfunctionalplatelets

2.1 Abstract....................................................17

2.2 Background.................................................18

vii

2.3 MaterialsandMethods

2.3.1 Characterizationof themicestudied.......................19

2.3.2 Productionofmaturemegakaryocytesexvivo...............20

2.3.3 Characterizationofexvivoderivedmegakaryocytes.........21

2.3.4 Isolationofplateletsandmegakaryocytesforinfusion........21

2.3.5 FlowcytometricstudiesininfusedhαIIb +mice..............22

2.3.6 InfusionstudiesinthrombocytopenichαIIb +mice...........22

2.3.7 Cremasterlaserinjuryfunctionalstudies...................23

2.3.8 FeCl 3carotidarteryinjuryfunctionalstudies................23

2.3.9 Infusedmegakaryocytefatestudies........................24

2.4 Results

2.4.1 Infusedmegakaryocytesandplatelets......................25

2.4.2 CharacterizationoftheinvivoderivedplateletsfromFLcells..26

2.4.3 FunctionalityofplateletsderivedfrominfusedFLcells.......37

2.4.4 Organdistributionstudiesofinfusedcells..................38

2.5 Discussion..................................................42

3. Platelettargetedprourokinaseasanovelthromboprophylaxisfibrinolytic

strategy

3.1 Abstract....................................................48

3.2 Background.................................................49

3.3 Materialsandmethods

3.3.1 AnalysisofantihαIIbmoAbs............................50

3.3.2 PlateletaggregationofantihαIIbmoAbs..................53

3.3.3 ConstructionandexpressionofantiPLTscFv/uPAT........53

3.3.4 BiochemicalcharacterizationofantiPLTscFv/uPAT........57

viii

3.3.5 BindingofantiPLTscFv/uPATtoh αIIb β3................58

3.3.6 InvitrofibrinolysisofantiPLTscFv/uPAT................59

3.3.7 EffectofantiPLTscFv/uPATinmodelsofvascular

thrombosis............................................59

3.3.8 FuturestudieswiththeantiPLTscFv/uPATfusionprotein...60

3.4 Results

3.4.1 BindinganalysisofantiplateletmoAbs....................63

3.4.2 BindinganalysisofantiplateletscFv......................63

3.4.3 AnalysisofantiplateletscFv/uPAT.......................67

3.4.4 FibrinolyticactivityofantiplateletscFv/uPATinvitro......70

3.4.5 Thrombolyticefficacy ofantiPLTscFv/uPATinmouse

Modelsofvascularthrombosis............................70

3.5 Discussion..................................................76

4. Summary,andfuturedirections....................................80

5. References......................................................85

ix

ListofTables Table21.Calculationofnumberofalveolicapillariesblockedbyinfused megakaryocytes............................................ 44 Table22.Supplementalmovielegend.................................. 47

x

ListofIllustrations Figure11.Megakaryopoiesisandplateletdevelopment.................... 5

Figure12.Plateletplugformationunderarterialcirculation ............... 8

Figure13.Fibrinolyticcascade ........................................ 10

Figure14.StructureofscuPAanditsvariouscleavedforms................ 13

Figure15.Depictionofthetwostrategiesproposed....................... 16

Figure21.IsolationandcharacterizationofFLcells...................... 27

Figure22.ComparisonofretroorbitalVstailveininfusionsofFLlargecells 28

Figure23.Flowcytometricdetectionanalysisofinfusedplatelets........... 29

Figure24.FlowcytometricanalysisofinfusedWTplatelets,andFLderived megakaryocytes........................................... 30 Figure25.FlowcytometricanalysisofinfusedWTplatelets,FLandBM derivedcellsonh αIIb +mice................................. 31 Figure26.FlowcytometricanalysisofinfusedWTplateletsandFLderived cellsinthrombocytopenicmice.............................. 32 Figure27.EffectofADAM17onthrombopoiesisfrominvitrogrownFLcells 34

Figure28.Characterizationoftheplateletsderivedfrominfusedcells....... 35

Figure29.CharacterizationofplateletsderivedfrominfusedFLcells....... 36

Figure210.Plateletincorporationintoarterialclotsafterlaserinjury....... 39

Figure211.FeCl 3carotidarteryinjurystudies........................... 40

Figure212.OrgandistributionstudiesofinfusedFLcells................. 41

Figure31.Finalconstructproposed.................................... 51

Figure32.MoleculardesignofuPAT.................................. 52

Figure33.DepictionofthestrategytogeneratetheantiPLTscFv.......... 54

Figure34.DepictionofthestrategytogeneratetheantiPLTscFv/uPAT.... 55

xi

Figure35.BindinganalysisofantihαIIbspecificmoAbstohumanplatelets.. 64

Figure36.PlateletaggregationanalysisofantihαIIbspecificmoAbs....... 65

Figure37.GenerationtheantiPLTscFv/uPATfusionconstruct........... 66

Figure38.ELISAanalysisofmoAbsandscFvinducedandnoninduced S2media................................................. 68 Figure39.CharacterizationofantiPLTscFv............................ 69

Figure310.CharacterizationoftheantiPLTscFV/uPATs................ 71

Figure311.FlowcytometricanalysisofhaIIbß3bindingbyantiPLT scFv/uPAT.............................................. 72 Figure312.BindingofantiPLTscFv/uPATtoimmobilizedh αIIb β3....... 73

Figure313.EnzymaticactivityofantiPLTscFv/uPAT................... 74

Figure314.ProphylacticthrombolysisbyantiPLTscFV/uPATinaFeCl 3 carotidarteryinjurymodel................................. 75 Figure315.Plateletandfibrinaccumulationinlaserinducedarteriole injuriesintreatedhaIIb +mice.............................. 77

xii

Chapter1

Introduction

1.1Background

1.1.1Pathogenesisofthrombosis

Thrombosis can occur in either the arterial or venous systems and both can be life

threatening.Acutearterialthrombosisistheproximalcauseofmostcasesofmyocardial infarction (heart attack) and of about 80% of strokes, collectively the most common causeofdeathinthedevelopedworld,andvenousthromboembolismisthethirdleading cause of cardiovascular associated death (1). Thrombosis manifests itself due to an imbalance in hemostatic procoagulant and anticoagulant mechanisms that maintain normalphysiologicalconditions.Theendotheliumisamajorcontributortomaintaining thisbalancebyprovidinganantithromboticsurfacethatinhibitsplateletadhesionand clotting. However, when the endothelium is perturbed by physical forces or by inflammation, the cells undergo programmatic biochemical changes that culminate in their transformation to a prothrombotic surface (2). During vascular injury, the coagulationsystem is activated withthe conversionof thrombinfromprothrombinby exposure of tissue factor. Thrombin is able to cleave fibrinogen to form fibrin, which contributestothestabilizationoftheplateletplug.Almostconcurrently,fibrinolysisgoes intoeffecttolimitandeventuallyremovethethrombusandrecanalizedthevessel.Itis importanttonotethatcoagulationandfibrinolysisaretwoantagonisticphenomenaand a balance is required amongst their respectiveactivatorsandinhibitorsforthe 1

processestobephysiologic(3).Thesemechanismsinvolvedinkeepingthehemostatic balancewillbediscussedinmoredetailinthesectionsthatfollow.

1.1.2Historyofplateletbiology

The discovery of platelets is best attributed to a summation of work by different scientists dating back to 1842, when platelets were recognized as little globules morphologicallydifferentthenredorwhitecells(4).Themostpowerfulevidence was provided by Giulio Bizzozero in 1881 and 1882, using intravital microscopy, he describedplateletsanatomicallyaswellastheiradhesivepropertiesupondamagetothe vascular wall (5, 6). Interestingly, 12 years before his platelet publication, Bizzozero observed large irregular cells in the bone marrow with highly lobulated, multilobed nuclei,buttheirsignificancewasunknownuntilWilliamHowellwithhis camera lucida studies in 1890 was able to corroborate on those observations and coined the term megakaryocytes(7). In 1906, Wright suggestedthat blood platelets were derived from thecytoplasmofmegakaryocytesnotingthesimilaritiesinbothcellswhenexposedtoa newpolychromestainingsolution(Wright’sstain)(8),andwiththesestudiesthebasic elementsofthrombopoiesiswereestablished.

1.1.3Megakaryocytedevelopment

Duringmegakaryopoiesis,committedhematopoieticstemcells(HSCs)undergoseveral stages of differentiation resulting in mature megakaryocytes that produce and release plateletsintothecirculation(9).HSCsarefoundprimarilyinthebonemarrow(10,11), andduringmammaliandevelopment,theycanalsobefoundintheembryonicyolksac, fetalliver,andspleen(12).Tostudymegakaryocyticprogenitors,invitroclonalassays were developed to analyze the cellular basis of lineage commitment and cell

2

differentiation(13,14).Basedonthesestudies,afewmegakaryocyteshavebeenshown

toariseinculture(Figure11)frommixedlineageprimitivecellsthataretermedcolony

forminguniterythroidmegakaryocyte(CFUGEMM)(15).More

differentiatedburstformingunitmegakaryocytes(BFUMeg)areabletoproducelarger

complex colonies that include satellite collections of megakaryocytes. The colony

formingunitmegakaryocyte(CFUMeg)canform350maturemegakaryocytesandare

the first fully committed cells to the megakaryocyte lineage base on their unique cell

surface markers (CD41 +Fc γlow CD34 +CD38 +CD9 ) (16). Subsequent cells are morphologicallyclosertoactualmegakaryocytesthentheirearliercounterparts.Thefirst oftheseimmediatemegakaryocyteprecursorsisthepromegakaryoblast,alsoknownasa stageImegakaryocyte.Itisnotabletoproducecoloniesinvitro,butabletomatureinto asinglemegakaryocyteinculture(17).Thehasadiploidnucleusanda diameterof1050 m,andthepromegakaryocyteorstageIImegakaryocyteis2080 m indiameterwithdevelopinggranulesinitscytoplasm(18).

Oncethemegakaryocyteprecursorsreachmaturity,theyhaveundergoneaseries of modifications including cytoplasm maturity, which involves the development of plateletspecificproteins,organelles,andmembranesystemsthatwillalsobepresentin platelets. They also undergo endomitosis, a process unique to megakaryocytes in mammals when they become polyploid by repeated cycles of DNA replication without undergoingcytokinesis(19).Thefinalmaturemegakaryocyteis50100 mdiameterand containsasinglelargepolyploidynucleus(2256N)(20).

Regulationofmegakaryopoiesishasbeenstudiedindetail,andithasbeenshownthat several cytokines are influential in maturation and development of megakaryocyte precursors.Themostinfluentialisthrombopoietin(TPO),apotentgrowthfactorthatis heavily involved in maturation and growth of early hematopoietic cells and the

3

megakaryocytelineage(21).Stemcellfactor(SCF),alsoknownaskitligand,hasbeen

showntobeinfluentialduringtheearlystagesofmegakaryopoiesis(22,23).Interleukin

6(IL6)and11arealsoinvolvedinregulationandseemtoworksynergisticallywithTPO andIL3(2426).

1.1.4Plateletdevelopment

Platelets are the final product of megakaryopoiesis and are essentially cytoplasmic portionsofmaturedmegakaryocytes.Inhumans,plateletshaveacirculatinghalflife of

710days,(27)andareproducedatasteadyrateof10 11 dailywithanincreaseofmore

then 20fold in times of heightened demand (15). It is estimated that one human

megakaryocytecanproduce~10 3plateletsand~10 2inmice(28).

Themechanismbywhichmegakaryocytesreleaseplateletsincirculationisstill

poorlyunderstoodinpartbecauseoftherarity(<0.1%)ofmegakaryocyteinnormalbone

marrow. Older studies attempted to explain the mechanismby proposing models that

focused on platelet budding from the surface of megakaryocytes and cytoplasmic

fragmentationviathedemarcationmembranesystem(2931).Bothofthesemodelshave

lostsupportduetoinconsistentobservations,andprimarilybecauseofthediscoveryof

TPO, which facilitated exvivo culture systems for the study of platelet biogenesis.

Currently the proplatelet formation model is more acceptable and is closely related to

Wright’s observation in 1906, that platelets were produced from long pseudopod

extensionsofmegakaryocytesthatthenreleaseplatelets(32).

Currentexvivostudieshavesupportedthatplateletsaremadefromnodesatthe

tipsofproplateletstrands(33),butthesesimplisticsystemslackthethreedimensional

organization, supportive cells and flow characteristics seen within the marrow. Direct visualization of live calvaria marrow using multiphoton intravital microscopy suggest

4

that

Figure11.Megakaryopoiesisandplateletdevelopment. HSCsundergoseveral stagesofdifferentiationuntiltheycommittoamyeloidprogenitorcell.TheCFUGEMM isofmixedlineagecapableofproducingmixedbloodcells.TheBFUMegisalsoableto produceamixtureofcellsaswellaslargemegakaryocytecolonies.TheCFUMegarethe first fully committed cells to the megakaryocyte lineage. Promegakaryoblast are consideredastageImegakaryocytefollowedbymegakaryoblastwhichhasadeveloped nucleusandthepromegakaryocytearestageIImegakaryocytewithdevelopinggranules in its cytoplasm. Shown are some of the cytokines involved at different stages of megakaryopoiesis.

5

that megakaryocytes release into the bloodstream large cytoplasmic fragments, not

small pseudoplatelets, that must be reorganized into mature platelets within the

organism (34). Indeed, older studies based on morphologic analysis of the lungs and

quantification of megakaryocyte like polyploidnuclei in thepulmonary venoussystem

suggestthatwholemegakaryocytestraveltothelungswheretheyreleasetheirplatelets

(3539).Thesepotentialmechanismsinvolvingplateletformationfrommegakaryocytes bycytoplasmicsheddingatthebonemarroworpulmonaryvasculaturebedsmayexplain whyithasbeenchallengingtoproducelargenumbersofplateletsfrommegakaryocytes

exvivo.

1.1.5Plateletroleinthrombusformation

Hemostasis is the process by which the body maintains the fluidity of blood under

physiologicalconditionsandpreventsexcessivebloodlossafterinjury(40).Plateletsare

essentialinbyformingamonolayerthatsupportsthrombingenerationand

platelet aggregation, contributing to stable thrombus formation. The degree of

involvement by platelets in a growing clot depends on the kind of circulation, for

example arterial thrombi form under conditions of high flow are composed mainly of

platelet aggregates bound together by thin fibrin strands. In contrast, venous thrombi

forminareasofstasisandarepredominantlycomposedofredcells,withalargeamount

ofinterspersedfibrinandrelativelyfewplatelets(41).

Under high shear arterial circulation, endothelial derived inhibitory factors

suppressplateletactivation,includingprostaglandinI 2 (PGI 2),andnitricoxide(NO)as wellasexpressionofCD39onendothelialcells(Figure12)(4244).Aftervascularinjury and exposure of subendothelium, a multistep complex process goes into effect, which involvesadiversearrayofextracellularmatrixcomponentsthatinteractwithplateletsto

6

formaplateletplug.Theplateletplugformationcanbedividedintothreestages(45), starting with the “initiation” step (Figure 12), when platelets roll on activated

endothelialcells,aprocessmediatedbyPselectinandEselectin(46,47).Theplatelets areabletobindtovonWillebrandfactor(VWF)viaitsGPIb/IX/Vmembranereceptor,

establishingatransientbondthatslowsthemdownandfacilitatetheiractivation(48).

TheplateletsalsoadheretoexposedcollageneitherdirectlyviatheGlycoprotein(GP)VI

and α2β1 integrin (49). This interaction promotes platelet activation, which leads to

morphologicalchanges,includingreleaseofgranularcontentandexpressionofactivated

GPIIb/IIIa,themajorplateletintegrinthatmediatesplateletaggregation.

Duringthe“extension”stage(Figure12),additionalplateletsarerecruitedwhen

activated platelets release agonists, such as adenosine diphosphate (ADP) and

thromboxane A 2 (TxA 2), which leads to further activation of adherent platelets. In addition tissue factor expressed at low levels by cytokine stimulated endothelial cells promotes local generation of thrombin (50). Thrombin is able to activate platelets through cleavage of the platelet Gprotein linked protease activated receptors (PAR1)

(51)aswellascleavageofGPVoftheGPIbIXVcomplex,whichallowsthrombintobind to GPIb α (52). Other surface molecules may also play an accessory role in platelet activationbythrombin.

The last stage of the platelet plug formation stabilizes the plug to prevent premature disaggregation. The stabilization is attributed to the close contact between activated platelets that allows contact dependent signaling (53). The final process of stabilizingtheplateletpluginvolvesthecoagulationcascade,whichisabletotransform soluble fibrinogen into insoluble fibrin by thrombin, thus forming the fibrinous meshwork.

7

Figure12. Plateletplugformationunderarterialcirculation. Plateletadhesion isstartedwhenVWFlinkscollagentoplateletsatsitesofinjury.Theactivatedplatelets release ADP, thromboxane and other chemicals that activate more platelets. Platelet adhesion occurs via the collagen receptors and the GPIb/IX/V complex followed by aggregation of additional platelets through GPIIb/IIIa, which forms a bridge through fibrinogentootheractivatedplatelets.

8

1.1.6Fibrinolysis

The fibrinolytic pathway has diverse roles including regulation of cellular activity and

tissuedevelopment,butitsprimaryfunctionsistheconversionofinactiveproenzymesto

activeformsforfibrindegradationandthusmaintainsbloodfluiditybybreakingdown bloodclots(54).Thecentralproteininthisprocessisplasminogen,whichisconverted byPAsintoplasminthatlysetheclot.ThevascularendotheliumisabletoreleaseuPA

andtissueplasminogenactivator(tPA)thetwoPAsinvolvedinthethrombolyticprocess.

PAsworktoactivateplasminogentoplasmin(Figure13),whichisabletolysefibrinby

attackingitatseveralsitesinthemolecule,leadingtofibrinclotlysis.Themechanismof activation by both PAs is by binding to specific cellular receptors, uPA binds to a

glycolipidanchormembraneproteinuPAR,andtPAtoatypeIItransmembraneprotein

(55).

Plasmin’sactivityiscontrolledbyanantiplasmininhibitor, α2antiplasmin,while tPAanduPAareregulatedbyformingcomplexeswithplasminogenactivatorinhibitor1

(PAI1),whichisfoundinbothplasmaandplateletagranules(56).Over90%ofPAI1is presentintheagranulesinaninactiveform,whichisthoughttobeoneofthereasons whyplateletrichthrombiareresistanttothrombolytictherapy(57,58).

1.1.7Keymediatorsoffibrinolysis

Plasminogenissynthesizedprimarilyintheliverandisfoundinhighconcentrationin plasma(59).Plasminogenisa92kDasinglechainglycoproteinconsistingof791amino acid(aa)residueswithGluastheNterminalaminoacid.Thestructureismadeupof five homologous kringlelike domains that mediate the interactions of the zymogen to regulatory sites, and cell surface receptors. Cleavage by PAs between Arg561Val562 converts plasminogen to the plasmin resultinginaheavychain(Achain)anda

9

light

Figure 13. Fibrinolytic cascade. Schematic cartoon of the fibrinolytic cascade.

Showninthecenterisafibrinclotatasiteofvascularinjury.Alsoshownisactivationof plasminogenbyPAstoproduceplasmin,whichisabletodegradethefibrinclot.

10

lightchain(Bchain)thatisenzymaticallyactive(60).

EndothelialcellssynthesizeandsecretetPAasasinglechainactiveenzymethat

circulatesinplasmaatrelativelowconcentrationswithahalflifeof5to7min(61).Itis made up of527 aa with a molecular weight(MW) of 70 kDa. tPA is composedoffive

domains;1)theNterminalregionof47residues(450aa)makethefingerlikedomain

homologous with fibronectin; 2) aa 5087 are homologous with the epidermal growth

factor(GFD);3)twokringleregions(aa87176and176262)whicharehomologouswith

thefivekringlesofplasminogen;and4)aserineproteinasedomain(aa276527)with

thecatalyticsiteHis 322 ,Asp 371 ,andSer 478 (62).ThehighaffinitybindingoftPAtofibrin

ismediatedbyboththefingerandkringletwodomains.ThisisimportantbecausetPAis

a poor enzyme in the absence of fibrin, but in the presence of fibrin, enhances the

activationrateofplasminogen(63).

uPAwasfirstisolatedfromurine,hencethenameurokinase.uPAissynthesized by vascular endothelial and smooth muscle cells, by epithelial cells, fibroblasts,

/, and also by multiple malignant tumors (6466). Unlike tPA

thatismainlyinvolvedinfibrinolysis,uPAhasadditionalfunctionsincellmigrationand

tissue remodeling (67). uPA is secreted as a single chain polypeptide (scuPA) with a

molecularweightof54kDathatconsistsof411aminoacidresidues(68).Thestructure

of uPA (Figure 14) is subdivided into three domains, starting with the Nterminal

domainhomologoustotheepidermalGFD(aa945)responsiblefortheinteractionof

urokinase with the uPAR/CD87 receptor. The kringle domain (aa 46143) contains a

sequence that interacts with the specific inhibitor PAI1. The Cterminal catalytic

domain, (aa 144411) includes the active site made up of serine proteases amino acid

triad His204, Asp255, and Ser356 (69, 70). Each domain of the protein has a rigid

structure supported by internal disulfide bonds, while intramolecular disulfide bonds

11

maintain the orientation of amino acid residues in the active center of the proteolytic

domain. This has been confirmed by loss of enzymatic activity after destruction or

irregularformationofdisulfidebonds(71).TheforminwhichsinglechainuPA(scuPA) issecretedhaslowintrinsicactivitybutcanbecleavedatseveralsitestomakeitactive.

Matrix metalloproteinase (Figure 14) (MMP3) specifically hydrolyzes the

Glu143 −Leu144peptidebondofscuPAandinitstwochain(tcuPA)derivative,yieldinga

17kDa NH 2terminal domain comprising uPAR’s binding site and a 32kDa COOH

terminalmoietycontainingtheserineproteinasedomainofuPA(72).Plasmincleaves

the Lys158Ile159 peptide bond producing fully active uPA as tcuPA, connected by

disulfidebonds.TherateofplasminogencleavagebytcuPAismorethen200foldhigher

thentherateofcleavagebyscuPA(73).ThisformofuPAlacktheGFPdomainsoitis

notabletobindtoitsreceptoruPAR,whichmakesitidealfortherapeuticstudiessinceit

eliminatesanysideeffectsthatresultfromreceptoractivationsuchasinflammation.

1.1.8ClinicaluseofPAsandtheirlimitations

One major form of treatment of occlusive thrombosis consists of pharmacological

dissolutionofthebloodclotbyintravenousinfusionofPAsthatactivatethefibrinolytic

system (74). Physical disruption of plaques (atherectomy) is also used as treatment

especiallyinperipheralarterialdiseaseofthelowerextremities(75).Thereareseveral

thrombolytic drugs in clinical use, including uPA and streptokinase that are effective

thrombolytics,butarenotfibrinspecific.Additionally,theyalsodecreasecirculatingand

intrathrombus plasminogen, potentially leading to reduction in clot lysis (76).

Furthermore, the bacterial derived streptokinase is immunogenic, resulting in drug

resistance,fevers,andallergicreactions(77).SecondgenerationagentssuchastPAand

scuPA are more fibrin selective and decrease the depletion of circulating plasminogen

12

and

Figure14. StructureofscuPAanditsvariouscleavedforms. scuPAstructureis

composedofthreedomains,theNterminaldomaincontainstheGFD,responsiblefor

theinteractionofuPAwithuPAR.Thekringledomaincontainsasequencethatinteracts withthespecificinhibitorPAI1.TheCterminalcontainsthecatalyticdomainwiththe

serineproteasesactivesite.scuPAcanbecleavedbydifferentcomponentstoformthe

activefortcuPA.MatrixmetalloproteinasecleavesbetweenGlu143 −Leu144andplasmin

cancleavebetweenLys158Ile159resultinginthetwochainderivatives.

13

and fibrinogen (78). All of these agents induce thrombolysis, and while there are

differences in fibrin binding affinity and other properties, in essence turn out to have

similarclinicalefficacyandsuchsideeffectsastheconsumptionofcirculatingfibrinogen

andplasminogenandthedevelopmentofbleedingcomplications(79).Theseagentsalso

have short halflives and require intravenous (IV) administration. Local infusions of

thesePAsatthesiteofpathologicthrombushavelimitedimprovedefficacy(8082).

1.2Dissertationobjective

Cardiovascular disease is estimated to be responsible for 17 million deaths per year,

makingitoneofthelargestcauseofmortalityworldwide(83).Currentantithrombotic

therapies reduce the risk of recurrent cardiovascular events, but increase the risk of bleedinginpartbecausetheyactsystemically.

Animprovedthrombolyticagentwouldhavebettertargetingofactivitytositesof

developingclots.Forthromboprophylaxisuse,anidealagentneedstoavoidtargetinga

maturethrombusandonlylysegrowingthrombi.Anadditionalusefulfeaturewouldbe

to have a therapeutic agent with a prolonged lifespan, preventing the development of

untoward thrombi over several days and allowing injured vessels to heal without

developing recurrent, pathologic thrombi. As proof of principle, our group has shown

thatuPAectopicallyexpressedinplateletsusingatransgenicapproachishighlyeffective

inpreventingarterialandvenousthrombosisandthattransfusionof5%oftheplatelet

mass protects WT mice from clotting without causing systemic fibrinolysis or

spontaneousbleeding(84).Basedonthesepreliminarydata,weenvisionachievinghigh

levels of megakaryocytespecific expression of uPA in human megakaryocytes derived

from in vitro differentiation of embryonic stem cells, allowing one to produce 14

modifieduPAcontainingplateletsfortargetingtositesofinjury(Figure15).Toreach

thatstage,wefirstneedtodevelopstrategiesthatfacilitatetheproductionofsuchdonor

derived platelets. One way to accomplish this is by infusing exvivo derived mature

megakaryocytesasasourcefordonorderivedplatelets.

Another plateletcentric strategy for producing effective thromboprophylaxis is bydevelopingarecombinantmodularPAthatspecificallybindstotheplateletsurface

andthatisactivatedonlyatasiteofactiveclotting(Figure15).Arecentstudytargeting

red blood cells confirmed the targeting efficiency, safety and efficacy for pulmonary

thromboprophylaxisusingamutatedlowmolecularweightscuPA,whichisspecifically

activatedbythrombin(85).Usingthesamethrombinactivatableproenzymefusedto

anantiplateletspecificscFvantibodyfragmentwouldlikelyprolongthehalflifeofthe

mutateduPAandprovideabettertargetingmechanismtositesofgrowingthrombi.The

thrombin generated from the growing thrombi serves as a control mechanism that

activates the uPAT, which can conceivably limit adverse effects, and increase its

duration in blood, prolonging its intravascular effect, and thus permitting use as

prophylaxis.

15

(A)

(B)

Figure 15. Depiction of the two strategies proposed. The two approaches proposedareshownstartingwith(A)theexpressionofuPAathighlevelsindeveloping megakaryocytes and produce platelets that will stored the zymogen in their alpha granulestobereleaseinatargetedfashionatsitesofinjury.Thesecondapproach(B) dealswithproducingachimericproteinthatwillbinddirectlytothesurfaceofplatelets viatheaIIbreceptorandwilldeliverathrombinactivatablelmwuPAtothethrombus sites,leadingtoclotlysisviafibrinolysis.

16

Chapter2

Infusingmaturemegakaryocytesintomiceyields

functionalplatelets

2.1Abstract

Thrombopoiesis,theprocessbywhichcirculatingplateletsarisefrommegakaryocytes,

remains incompletely understood. Prior studies suggest that megakaryocytes shed

platelets in the pulmonary vasculature (35, 36, 38, 39, 86). To better understand

thrombopoiesis and to develop a potential platelet transfusion strategy that is not

dependentupondonors,weexaminedwhethermegakaryocytesinfusedintomicesheda

significantnumberofplatelets.Wefoundthatinfusedmegakaryocytesledtoclinically

relevant increases in platelet numbers. The released platelets were normal in size,

displayedappropriatesurfacemarkers,andhadanearnormalcirculatinghalflife.The

functionalityofthedonorderivedplateletswasalsodemonstratedinvivo.Theinfused

megakaryocytesmostlylocalizedtothepulmonaryvasculature,wheretheyappearedto

shedplatelets.Thesedatasuggestthatitmaybeunnecessarytogenerateplateletsfrom exvivo grown megakaryocytes to achieve clinically relevant increases in platelet

numbers.

17

2.2Background

In2001,platelettransfusionsintheUSAtotaled10,196,000units,anincreaseof12.6%

from 1999 (87). While the need for healthy platelet donors is increasing, there is a

significant donor shortage due to the growing population of patients with serious

medical illnesses associated with thrombocytopenia and hemorrhages. Platelets for

transfusionsalsosufferfromthefollowingconcerns:variabilityofdonorunitsinterms ofnumberofplateletsandfunctionality,riskofdonortopatientinfectioustransmission, theshortlifespanofroomtemperaturestoredplatelets(necessarytomaintainplatelets

ofnormallifespan),bacterialcontaminationduringthisstorage,andthedevelopment

ofplateletalloantibodiesinmultiplytransfusedpatients.Alloftheseproblemshighlight

aneedfornewstrategiestogenerateplateletsforinfusiontherapy.

Two models of thrombopoiesis have been proposed: The release of platelets

producedfrommegakaryocyteswithinthemarrowspaceandthereleaseofplateletsby

circulating megakaryocytes within the pulmonary vasculature (39). Invitro studies

suggested that platelets form nodes at the tips of proplatelets strands (33). However,

direct visualization of live calvaria marrow using multiphoton intravital microscopy

suggestedthatmegakaryocytes,adjacenttothevascularspace,releaselargecytoplasmic

fragments into the bloodstream (34). These must then be reorganized into mature

platelets.Olderstudiesbasedonmorphologicanalysisofthelungsandquantificationof

megakaryocytelike polyploid nuclei in the pulmonary venous system suggested that

megakaryocytestraveltothelungswheretheyreleasetheirplatelets(35,36,38,39,86).

Final platelet formation from megakaryocyte cytoplasmic shedding at the

marrow/endothelial interface or in the pulmonary vasculaturebeds may explain 18

whyithasbeenchallengingtoproducelargenumbersofplateletsfrommegakaryocytes

growninvitrowithoutsimulatingspecialnichesand/orflowconditions.

Derivationofplateletsfrommegakaryocytesinculturewasfirstreportedin1995

(88),buthasbeendifficulttoquantitativelyupscale.Todatethebestpublishedresult from infused invitro produced platelets used irradiated mice with lowplatelet counts

(~10 4/l)(10).Eveninthissetting,peakdonorplateletcountswere1%2%ofthislow numberofplateletsandthisminisculeincreaseisunlikelytobeofclinicalutility.Given the limited success with which platelets have been generated ex vivo, we examined whetherinfusedmegakaryocytesreleaseplateletsinvivo.Wefoundthatbyinfusingex vivo generated megakaryocytes into mice, we can achieve approximately an 100fold increase in recipient platelet count over the prior best published results, thereby reachingclinicallyrelevantlevelsofdonorplatelet.Theseplateletshaveaslightlyshorter halflife than infused platelets, but are normal in size and surface markers and are functionalintwoclottingmodels.Wealsoshowthattheinfusedmegakaryocytesappear tobetrappedinthepulmonarybedwheretheyshedtheircytoplasm.

2.3Materialandmethods

2.3.1Characterizationofthemicestudied

DonorcellsandplateletswerederivedfromC57BL/6WTmice(JacksonLaboratory)or mUK transgenic mice that ectopically expressed murine urokinase within megakaryocytesandthathadbeenbackcrossedtotheC57BL/6background>10times

(84). The recipient hαIIb + mice were previously described (89), and are homozygous transgenicforh αIIb(h αIIb +/+ )thatarealso null for the expression of platelet m αIIb 19

(m αIIb /).Thesemiceplateletsexpressh αIIblevelsat~20%ofthelevelseenonhuman plateletsandarefunctional(90).mUKandh αIIb +miceweregenotypedbypolymerase chain reaction (PCR) analysis of tail genomic DNA using previously described primer pairsandreactionconditions(84,89).AllanimalstudiesweredonewithInstitutional

AnimalUtilizationCommitteeapprovalattheChildren’sHospitalofPhiladelphia.

2.3.2Productionofmaturemegakaryocytesexvivo

Moststudiesweredonewithfetalliver(FL)derived megakaryocytes, beginning with

E14 FLcells that were homogenized to a single cell suspension by sequential passage through18,23,and25gaugeneedlesaspreviouslydescribed(91).Thecellsuspension was passed through a 100mnylon mesh filter and cultured in Iscove’s Modified

Dulbecco’s Medium (IMDM) with 10% calf serum, 2 mM Lglutamine, 1% penicillin/streptomycinand50ng/mlofrecombinantmurineTPO(R&DSystems).After

10daysinculture,over90%oftheresultantcellsweremegakaryocytesasdeterminedby acetylcholinesterasepositivity(datanotshown).Thematuremegakaryocytepopulation was isolated using a previously described 2stage velocity sedimentation method for isolating megakaryocytes (92), which involves centrifugation through a 1.5% and 3% bovineserumalbumin(BSA)gradientat200RPMsfor30min.

Adultbonemarrow(BM)cellswereobtainedfromfemursandtibiaeofC57BL/6 miceaspreviouslydescribed(93).Thecellsfromthemarrowlumenwereflushedwith sterileDulbecco’sPhosphateBufferedSaline1X(PBS)(Invitrogen)usinga10mlsyringe and a 25gauge needle. The cells were disrupted and passed through a 100mnylon mesh filter. They were then cultured in the same media as the fetal liver cells and isolatedafter8daysofcultureinthesamemanner.

20

2.3.3Characterizationofexvivoderivedmegakaryocytes

FL and BMmegakaryocytes to proplatelet number was determined visually with a

hemocytometer (94).DNAploidyof the derived megakaryocytes was assessedby flow

cytometryaspreviouslydescribed(95).IsolatedpopulationswerewashedwithPBSand

stained with 10g/ml monoclonal fluorescein isothiocyanate (FITC)labeled anti

murineCD41antibody(BDBioscience)for30min.Thecellswerethenrinsedtwicewith

PBSandresuspendedinpropidiumiodidesolution(0.1%sodiumcitratewith50g/ml

of propidium iodide) (Sigma) for 4 hrs. The cells were then incubated with 10 U/ml

RNaseA(Sigma)for90minatroomtemperature(RT)andanalyzedbyflowcytometry

usingaFACScan(BectonDickinson).

2.3.4Isolationofplateletsandmegakaryocytesforinfusion

WashedplateletsfromwholebloodwereisolatedfromtheinferiorvenacavaofC57BL/6

mice under direct observation and collected into acid citrate dextrose (ACD) as

previouslydescribed(96).Plateletrichplasmawascollectedaftercentrifugationat200 g

for5minatRT,andprostaglandinE1(Sigma)wasaddedtoafinalconcentrationof1

mM.Plateletswerethenpelletedbycentrifugationat800 g for10minatRT.Thepellet waswashedincalciumfreeTyrode’sbuffer(134mMNaCl,3mMKCl,0.3mMNaH 2PO 4,

2mMMgCl 2,5mMHEPES,5mMglucose,0.1%NaHCO 3,and1mMEGTA,pH6.5), andresuspendedinCATCHbuffer(PBS,1.5%BSA,1mMadenosine,2mMtheophylline,

0.38% sodium citrate) (all from Sigma). Platelet counts were determined using a

HemaVet counter (Triad Associates), and the appropriate amount of platelets was infusedintorecipientmice.Megakaryocyteswereproducedasdescribedin“Isolationof plateletsandmegakaryocytesexvivo”.Oncethematurepopulationofmegakaryocytes was obtained from the BSA gradient, the cells were washed in CATCH buffer and

21

countedwithahemocytometerandinfusedeitherretroorbitallyorbytailvein.

2.3.5FlowcytometricstudiesininfusedhαααIIb +mice

Before infusion of cells or platelets, whole blood was collected retroorbitally from recipient mice into a mini capillary blood collection tube (RAM Scientific) and again afterwardsattimepointsextendingfrom5minto36hrs.Thebloodwasdoublestained with monoclonal FITCconjugated mouse, antihumanspecific CD41 antibody

(eBioscience) and monoclonal phycoerythrin (PE) rat, antimouse CD41specific antibody(BDBioscience)for30minandanalyzedbyflowcytometryusingaFACScan.

Activationoftheinfusedplateletswasassessedby3colorwholebloodflowcytometry, usingamonoclonalmouseantihumanCD41labeledwithperidininchlorophyllprotein conjugatedwithacyaninedye(PerCPCy5.5),PEratantimouseCD41,andmonoclonal

FITCratantimouse Pselectin. To examine the relative expression of membrane receptorsinrecipientversusdonorplatelets,wholebloodwasstainedwithmouseanti humanCD41(PerCPCy5.5),PEratantimouseCD41,andeithermonoclonalantibodies

FITClabeledratantimouseGPIb αorratantimouseGPIX(EmfretAnalytics).

2.3.6Infusionstudiesinthrombocytopenich αIIb +mice

Miceweresubjectedtoahighdoseofirradiation(1,000centigraystotal;twosessions,24 hours apart). Platelet counts were initially monitored daily to determine a temporal platelet profile. Animals included in these studies had platelet counts between 12 x

10 8/mlonday7afterirradiationandimmediatelybeforeinfusionofthecellsin200 l

CATCHbuffer.Additionalplateletcountswereperformedat4,24,and48hoursafter infusion.

22

2.3.7Cremasterlaserinjuryfunctionalstudies

WashedplateletsandisolatedFLderivedmegakaryocyteswereinfusedintohaIIb +male

mice. One hour after infusion, the mice were anesthetized using intraperitoneally

injected sodiumpentobarbital (11mg/kg, Abbott Laboratories), and maintained under

anesthesiawiththesameanestheticdeliveredviacatheterizedjugularveinasdescribed

(97). Fab fragments from the antimouse CD41 antibody were produced using the

ImmunoPureFabPreparationkit(Pierce Biotechnologies)andconjugatedwithAlexa 488

using an Alexa Fluor Protein labeling kit (Molecular Probes). These labeled fragments were injected intravenously into the cannulated jugular vein 5 min before injury. The cremastermusclewasisolatedandthemicrovesselsstudiedusinganOlympusBX61WI microscope (Olympus) with a 40X/0.8 numeric aperture (NA) water immersion objective lens. Laser injury was induced using an SRS NL100 Nitrogen Laser system (Photonic Instruments) at 65% energy level with blood vessels ranging in size from 20 to 40 m. A visual confirmation of small extravasations was made for each studiedbloodvesselasanassurancethattheinjurywasmadeaswellasanindicatorof consistentinjury.Datawascollectedoveracourseof2.5minat5framespersecond(f/s;

750framesperstudy).Nomorethan5arterioleand5venuleinjuriesweredoneineach mouse.

2.3.8FeCl 3carotidarteryinjuryfunctionalstudies

FeCl 3induced arterial injury was performed as previously described (98) with minor changes.HαIIb +micewereanesthetizedbyintraperitonealinjectionofpentobarbital(80 mg/kgNembutal;OvationPharmaceuticals)approximately1hrafterinfusionofeither

WTormUKmegakaryocytes.Thecarotidarterywasisolatedbybluntdissection,anda

Doppler flow probe (Model 0.5VB; Transonic Systems) was positioned the

23

aroundvessel,followedbya1x2mm 2pieceoffilterpaper(#1;WhatmanInternational) soakedin20%FeCl 3thatwasplacedonthearteryfor3min.Theareawasthenflushed withPBS,andbloodflowthroughthearterywasmonitoredfor30min.Totalflowand

timetothefirstcompleteocclusionof>10minwererecorded.

2.3.9Infusedmegakaryocytefatestudies

BrdU studies were carried out to better understand the fate of the infused

megakaryocytes. FL cells were cultured for 5 days and afterwards exposed to 10 M

BrdU(Sigma)intheirgrowthmediafor2moredays.Thecellswereisolatedandinfused

into hαIIb + mice. The mice were sacrificed, and organs, including brain, heart, liver, spleen,bonemarrowandlungsweredissectedat30min,6hrs,and24hrs.Thelungs were flushed with PBS and inflated with 10% formalin solution (Sigma) to preserve alveolar architecture. All organs were fixed in formalin overnight at 4°C for immunohistologicalanalysis.DetectionofBrdUlabelednucleiwasperformedwitharat polyclonal antiBrdU antibody diluted 1/50 (v/v, Abcam) and a biotinylated rabbit polyclonalantiratIgGdiluted1/200,wasusedasthesecondaryantibody.Detectionof infused megakaryocyte cytoplasm in the lungs was performed by labeling with a goat polyclonalantibodyagainstmurine αIIb(SantaCruzbiotechnology)andabiotinylated

rabbit polyclonal antigoat IgG antibody, diluted 1/200 as the secondary antibody for

detectionwasdoneusingaDAKOkit(EnVision).

24

2.4Results

2.4.1Infusedmegakaryocytesandplatelets

To address whether infused megakaryocytes give rise to circulating platelets, we

generated FL and BMderived magakaryocytes (Figure 21 A). This strategy has been

described to yield a large number of mature megakaryocytes (91). Mature

megakaryocyteswereseparatedfromothercellsandmostoftheproplateletsbyatwo

stagevelocitysedimentationtoproducea“largecell”fractionwithoverhalfofthecells

having a diameter >50 m and with only ~2.5:1 proplatelets:cell (Figure 21 B). The

remaining“smallcell”fractionwas>95%smallmegakaryocytesandproplateletswith

~10:1proplatelets:cell(Figure21B).Ploidyanalysiswasperformedonbothfractions and showed that the small cell fraction had low DNA ploidy relative to the large cell fraction(Figure21C).

WTplateletswereusedaspositivecontrolsforstudiesinwhichoneofthetwo cellfractionsofexvivogrownFLandBMmegakaryocyteswereinfusedintoh αIIb +mice eitherretroorbitallyorbytailvein;thetwoapproachesgavesimilaroutcomes(Figure2

2).Therecipientanimalshaveonlyh αIIbontheirplateletsurface(89),whichallowsfor flowcytometricdetectionanalysisoftheinfusedWTplateletsusingspeciesspecificanti

αIIb (CD41) antibodies (Figure 23). After infusion, blood derived WT platelets were detected in h αIIb + recipient mice immediately at the 5 min time point (Figure 24 A) withahalflifeofapproximately36hours(Figure25A).

The donor platelets were present in recipient blood as expected based on the initialplateletcountoftherecipientmouseandanestimatedbloodvolumeof2ml(data notshown).Afterinfusingthelargecellfractions,donorderivedplateletswereseen,but with delayed kinetics: only a few donor platelets were detectable at 5 min, 25

followed by a peak at approximately 90 min (Figure 24 B and Figure 25 A). These

platelets had a slightly shorter halflife of approximately 20 hrs than when donor plateletshadbeeninfused.Basedonthenumberoflargecellsinfused,thepeakincrease in platelet count and a recipient mouse blood volume of 2 ml, we calculate that each

large cell gave riseto100200 platelets, assuming all large cells gave rise toplatelets.

Infusedsmallcells,whichwerericherinproplatelets,gaverisetoanimmediatepeakjust

asinfusedWTplatelets;however,theseplateletshadasignificantlytruncatedhalflifeof

approximately2hrs(Figure25A).

InfusingadultBMmegakaryocytesgrownincultureinthepresenceofTPOand

fractionated as in Figure 21 B, we were also able to detect circulating platelets. The

numberof plateletspermegakaryocytewassimilar to FLderived megakaryocytes, the

halflifewasslightlylongerat24hrs,buttheyieldofmegakaryocytesfromadultmarrow was lower compared to FL cells. Consequently, further studies below focused on FL

derivedmegakaryocytes.

To simulate clinical thrombocytopenia,we irradiated mice and infused CATCH buffer,WTplatelets,orFLlargecellsneartheinducedplateletnadirandobservedthat bothplateletsandmegakaryocytessignificantlyincreasedtheplateletcountrelativeto

CATCHbufferoveratimecourseofmorethan24hours(Figure26).

2.4.2CharacterizationoftheinvivoderivedplateletsfromFLcells

Plateletsderivedfromthelargecellfractionhadashorterhalflifethaninfusedplatelets.

Thesmallcellfractionhadanevenmoretruncatedhalflife.Wewouldhaveanticipated thattheplatelets,especiallythosefromthelargecellfraction,(withmostlynewlyformed platelets) would have a longer halflife than infused washed platelets. One clear possibilityofwhyFLderivedlargecellplateletshadashortenedhalflifecouldbethat

26

these

Figure21.IsolationandcharacterizationofFLcells .(A)Thestrategyfollowed in our study is presented by which the three different products for infusion were collected:isolatedWTplatelets,andthelargeandsmallcellfractionsobtainedfromFL cells grown in the presence of thrombopoietin (TPO). (B) Representative fields of the small and largecell fractions.Scale bars: 100 m.(C)Representative analysis of DNA

27

contentofthesmallandlargecellfractions.

Figure 22. Comparison of retroorbital Vs tail vein infusions of FLlarge cells . Flow cytometric percentage of infused ~10 6 FLlarge cells in recipient animals.

N=5studiesperarm.Mean±1standarddeviation(SD)areshown.

28

Figure 23. Flow cytometric detection analysis of infused platelets. Flow

cytometricanalysiswasperformedonwholebloodfromhaIIb +recipientmiceinfused with WT donor platelets. Gate 1 (platelets) was set up in the forward scatterH/side scatterH plot (A) representing the overall platelet population. Gate 2 included either recipient (antihaIIb + FITC) platelets (B) or donor (antimaIIb + PE) platelets (C). The percentage of the recipient versus donor platelets was calculated from the FL1xFL2 densityplotthatincludedcellssatisfyingbothGates1and2,asshownin(D).

29

Figure24.FlowcytometricanalysisofinfusedWTplatelets,andFLderived

megakaryocytes. Flowcytometricanalysisonwholebloodfromh αIIb +recipientmice beforeandatdifferenttimepointsafterretroorbitalinfusionofdonorcells.Bloodwas stainedwithlabeledspeciesspecificantiαIIbantibodies.The%ofplateletsintheleft upper quadrant represents the recipient’s platelets, while the % in the right lower quadrantrepresentsdonorderivedplatelets.(A)Flowcytometryfromrecipientmouse before and after infusion of 10 8 platelets (positive control). (B) Flow cytometry from recipient mouse before and after infusion of 10 6 FLderived large cells.

30

Figure 25. Flow cytometric analysis of infused WT platelets, FL and BM

derivedcellsonhαααIIb +mice.(A)Flowcytometricpercentageofinfused10 6FLlarge

cellsand(B)infused10 6adultBMcells.N=5forWTplatelets,9forFLcells,5forBM studies.Mean±1SDareshown.

31

Figure26.FlowcytometricanalysisofinfusedWTplateletsandFLderived cells in thrombocytopenic mice. Percent platelet rise in irradiated thrombocytopenic mice postinfusion. N=5 per arm. Mean ± 1 SD are shown. Initial

plateletcounts(10 8/ml)inthethreegroupswere:CATCHbuffer=1.8±0.2;platelets=

1.9±0.3;andlargecells=1.0±0.2.

32

thesearefetalplateletswhichtendtohaveashorterhalflifethenadultplatelets(99).To seeifthehalflifeimproveswhenusinganadultsourceofmegakaryocytesinsteadofour

fetalliverderived,weinfusedculturedadultBMmegakaryocytes,whichhadahalflife

perhaps as good as infused WT platelets,but clearly not longer (Figure 25 B). Prior

studieshavesuggestedthatametalloproteinase,tumornecrosisfactoralphaconverting

enzyme(ADAM17),couldpotentiallybepresentinthemediaofculturedmegakaryocytes

andberesponsiblefortheshorterhalflife,butgrowingthecellsinthepresenceofeither

metalloproteinase inhibitors GM6001 (Ilomastat) or TAP1 did not improve the half

lives(Figure27).

OneindicatorofplateletactivationwouldbethepresenceofdonorWTplatelet

microparticles.However,thesizedistributionoftheplateletsderivedfromthelargecell

fractionwasidenticaltothatseenforinfusedWTplatelets(withoutasignificantpoolof

microparticlesbyforwardscatter)(Figure28).Anotherindicatorofcellactivationisthe

expressionofPselectinontheplateletsurface(100,101).Priorstudiesinvolvingplatelet

concentrate storage and transfusion have shown that this marker is increased on

activatedplatelets(102).Flowcytometricanalysisdidnotsupportplateletsderivedfrom

infused megakaryocytes having a higher level of surface Pselectin expression than

infusedcirculatingplatelets(Figure29A).Moreover,ADAM17cleavestheglycocalicin

extracellular portion of GPIb α, inactivating the GPIb/IX receptor without altering surfacereceptordensity(103,104).Wemeasuredrelativeexpressionoftheextracellular portionofGPIb α(Figure29B)versusGPIX(Figure29C)onplateletsderivedfromthe

infused large cell fraction as well as from donor washed platelets and found no

difference. Thus, it appears that the shortened halflife of the platelets derived from

infusedmegakaryocytesmightnotbeduetotheeffectsofthepreviouslydescribedex vivometalloproteinaseADAM17.

33

Figure 27. Effect of ADAM17 on thrombopoiesis from invitro grown FL

cells. WTFLcellsweregrownincultureinthepresenceofeither100MGM6001(A

and B)or 10 MTAP1(C andD) and then separated into small and large cells asin

Figure22(B),andinfusedintoh αIIb +recipientmicetoseeiftheseADAM17inhibitors wouldimprovethehalflifeobservedofplateletsderivedfrominfusedlargecells.N=2,

performedinduplicates.

34

Figure28.Characterizationoftheplateletsderivedfrominfusedcells .(A)

Sizedeterminationofcirculatingdonorplateletsfrominfusedplateletsor(B)smallcell

or (C) large cell fractions. Shown is a representative study of the forward versus side

scatter of the whole blood samples measured with the two αIIb positive populations highlightedinbluetherecipientplateletsandinredfortheinfusedplateletsorderived platelets. 35

Figure 29. Characterization of platelets derived from infused FLcells . (A)

Representative flow cytometric analysis of Pselectin on infused donor cells resulting fromeitherinfusedplateletsorcellfractions.(B)RepresentativerelativelevelsofGPIb α extracellular domain to (C) GPIX in donor platelets from infused platelets or cell fractions.

36

2.4.3FunctionalityofplateletsderivedfrominfusedFLcells

Clinically, platelets are transfused not to increase platelet number, but to reverse or

preventableedingdiathesisduetotheabsenceofplatelets.Wehaveusedtwodistinct

assaystolookatplateletfunctionafterinfusedmegakaryocytes.Inthecremasterlaser

injury model, we looked at the incorporation of both infused platelets and platelets

derivedfrominfusedlargeorsmallcellsintogrowingclotsafterlaserinjurytocremaster

arterioles.Inallthreesettings,donorderivedplateletsreadilybecomeincorporatedinto

thedevelopingthrombianddidsoatleasttothesameextentasforWTdonorplatelets

(Figure210AandB).

Inclusion of platelets derived from the small cells into growing clots was less vigorous.Moreoverinthissetting,therewasadistinctpopulationofCD41 +cellsthatre

circulated, rarely becoming incorporated into a thrombus (Figure 210 C and D) and

SupplementalVideo1).Thisphenomenonwasrarelyseenafterinfusionofthelargecell

fraction (Figure 210 B) and Supplemental Video 2), and never with infused platelets

(Figure210A),andSupplementalVideos3).

WealsoexaminedplateletfunctionusingaFeCl 3carotidarteryinjurymodelafter infusingeitherCATCHbufferorFLWTlargecellsintotheh αIIb +recipientmice.These

recipientmicehaveamildbleedingdiathesisinpartbecauseofthepresenceofonly20%

ofthedensityofCD41comparedtoWTmice(89).WefoundthattheinfusedWTlarge

cellsleadtoashortenedtimetodevelopmentofstableocclusion(5.3±0.5versus7.4±

0.9likelyduetothepresenceofWTdonorderivedplatelets.Wealsoinfusedlargecells

grown from the FLs ofmice that ectopically express urokinase and store it in their α

granules(mUKmice)similartopatientswithQuebecPlateletDisorder(105).Wehave

already demonstrated that mUK mice are resistant to thrombosis in the FeCl 3 carotid

artery injury model and mUK platelets transfused into a WT mouse block clot

37

development(84).WereasonedthatifinfusedlargecellfractionderivedmUKplatelets

are functional then they would interfere with clotting in the h αIIb + recipient mice.

Indeed,h αIIb +miceinfusedwithmUKlargecellsfailedtoformstableocclusions(Figure

211A),andtotalbloodflowoverthe30minstudywasmarkedlygreaterinthemUKcell

infusedmicethaneithercontrolgroups(Figure211B).

2.4.4Organdistributionstudiesofinfusedcells

The above studies are consistent with the concept that mature megakaryocytes get

trapped in the pulmonary vasculature and shed functional platelets. Smaller and less

mature megakaryocytes may be able to escape the pulmonary capillary bed and

recirculateorgettrappedinotherorgans.Tobegintoaddressthismodelofevents,we

labeled the nuclei of maturing cells in culture by the inclusion of 5bromo2

deoxyuridine (BrdU) in the growth media prior to infusion of the cells, and isolated variousorgansatintervalsrangingfrom30minupto36hrs.Themajorstainingwasin

thelungs,andthiswasmoreintenseafterinfusionofthelargecellfraction(Figure212

A).Stainednucleiweremaximalatthefirsttimepointcheckedandwerevisibleforupto

24hrs,butnotat36hrs(Figure212B).Afewnucleiwerevisibleintheredpulpofthe

spleenafterbothsmallorlargecellfractioninfusions(Figure212C).Nonewerepresent

intheliver,heart,brainorbonemarrow(datanotshown).

AdditionallytheBrdUlabeledmegakaryocytesappeartoretaintheircytoplasm

forupto30minpostinfusioninthemicrovesselsofthelungs(Figure212BandD).

This observation would be consistent with the delay seen in peak platelet count after

infusedlargecellfractionasseeninFigure24(B)andFigure25(A).Toconfirmthis

observation, we stained the tissues with an antimurine CD41 antibody and detected

positivestainingcytoplasmpresentupto30min(Figure212D).

38

Figure210.Plateletincorporationintoarterialclotsafterlaserinjury .(AC)

Representative images of platelets incorporating into clots after infused platelets or

indicatedcells.(A)Donorplateletsdetectedusinga labeledantimαIIbantibody.(B)/(C)

Same as (A), but after large and small cells infused, respectively. (D) Sequential stills fromlefttorightnotingarecirculatingm αIIb +cell(arrowhead)aftersmallcellinfusion.

(E)Summationofdonorplateletsincorporatedintogrowingthrombiaftereitherinfused

platelets or cells. Twenty movies were evaluatedpergraph. 39

Figure211.FeCl 3carotidarteryinjurystudies .(A)Timetostablecompletevessel occlusionafterinfusedCATCHbufferorWTlargecellsormUKlargecells.(B)Sameas in(A),butmeasurementoftotalflowoverthe30minstudy.For(A)and(B),N=5per arm.Mean±1SDshownfor(B).

40

Figure212.OrgandistributionstudiesofinfusedFLcells .(A)Stainingoflung fromhaIIb +miceinfusedwithPBSsmallorlargecellsgrowninBrdU(arrowspointto stainnuclei).(AC)Originalmagnification=50X, scalebar=200 m.(B)Kineticsof

BrdUlabeled large cells in the lungs.(C)Same as (A) but for spleen. (D) Highpower

(200X) of lungs: BrdUlabeled nuclei (left) andm αIIb(right) 30 min postinfusion of large cells. Scale bar = 50 m. Data are representativeof3separatestudies. 41

2.5Discussion

Thrombopoiesis is a unique phenomenon developed in mammals to have circulating

anuclear subcellular fragments that can participate in clot development when the

circulatorysystemisperturbed(106).Inheritedoracquireddisordersofmegakaryocyte differentiation or cytoskeletal maturation can affect thrombopoiesis and final platelet counts(107110).Understandingthrombopoiesisisimportantifoneistodevelopexvivo methodologiesfortheproductionofclinicallyrelevantnumbersofplatelets.

Whilemuchhasbeenlearnedaboutcytoskeletalcomponentsinthrombopoiesis,

it is still unclear where platelets are made and how megakaryocytes shed them. The

methodology used to study thrombopoiesis has to a large extent defined the potential

underlying mechanism. Studies of maturing megakaryocytes in culture suggest that a

mature megakaryocyte forms an interconnected network of “beadsonastring”

proplateletsthatcouldeventuallybecomeplatelets(33).Thismodelwouldsupportin vivo thrombopoiesis localized within the marrow space itself. Instead, multiphoton

intravital microscopy of marrow space suggests that megakaryocytes adjacent to the vascularliningshedlargecytoplasmicfragmentsthatmustthensecondarilyreleasefinal

platelets within the vasculature (34). Our studies and prior ones that included

pulmonary histology (35, 36, 38, 39, 86) suggest a third mechanism wherein normal

sized and functional platelets are released from circulating, mature, highploidy

megakaryocytes directly within the vascular bed. These models are not necessarily

mutually exclusive: Large cytoplasmic fragments may be the norm, while whole

megakaryocytesarelessfrequentlyreleasedintothecirculation.Bothlargecytoplasmic

fragments and whole megakaryocytes may shed platelets within the pulmonary bed

using mechanisms consistent with the in vitro studies, but with concurrent flow 42

andlocalvascularfactorsinvolvedasothershaveproposed(111).

Intuitively, one would expect that megakaryocytes lodging in the lungs might

poseacardiovascularchallenge.However,ifamousehas12X10 9plateletsandthese have a halflife of 5 days (112), and if a megakaryocyte sheds 10 2 platelets, then one wouldneed24X10 6 megakaryocyteseachdaytravelingtothelungsto maintainthe animal’splateletcount.Weinfused10 6megakaryocytesinmostofourstudies,andwhile

deathasanendpointwasnotmeasured(allarmsofourstudieshadsomeanimallosses, but no one arm was particularly harmful), infusing what is near to a daily number of

neededmegakaryocytesovera10minperiodwastolerated.Sincethemousehas2.3x

10 6 alveoliinbothlungs(26),andeachalveolusistraversedbymanycapillaries(25100; based on human alveolar structure, corrected for the difference in volume between human and murine alveoli (113), our calculation is that each mouse lung contains approximately10 8capillaries,whichiswellwithintheestimatesof4x10 11 capillariesfor

the human lung (113). Given this number of capillaries, we anticipate that we are blocking~1%ofpulmonarycapillariesforupto24hrsfromourkineticstudiesinFigure

211(Table21).

Inadditiontothepulmonaryvasculaturehavingthecapacitytohandleaninflux

of megakaryocytes, another question is whether the pulmonary vasculature offers a

special niche for platelet release. Wouldmegakaryocytes get trapped inother vascular beds and if so would they be as efficient in thrombopoiesis? Clearly a few

megakaryocytes reached and were trapped in the splenic red pulp, but whether these

megakaryocytescanshedplateletsisunclear,eventhoughsomesuggestthatthisisalso

a site for platelet production (114). Individuals with significant right to left

cardiovascular shunts are known to have lower platelets counts, which others have

proposedtobeduetopulmonarybypassbycirculatingmegakaryocytes(115).However,

43

Table 21. Calculation of number of alveoli capillaries blocked by infused megakaryocytes. Miceplateletcontent:12X10 9platelets(112) Miceplatelethalflife:37hrs(118) Megakaryocytesheds10 2platelets(determinedinthisstudy). Calculate: 2-4 X 10 6 megakaryocytes each day traveling to the lungs to maintain the animal’s platelet count. Micealveoli:2.3X10 6inbothlungs(119) Capillaries per alveoli: 25100; based on human alveolar structure, corrected for the differenceinvolumebetweenhumanandmurinealveoli)(113) Calculate: Infusion of = 10 6 megakaryocytes over 10 mins into the mice will block 0.4- 1.7% of the entire capillary bed for a 24 hr period.

FLderivedmegakaryocytesthatnaturallybypassthepulmonarybedcanformplatelets

(116, 117), suggesting that other vascular beds may be able to substitute for the

pulmonaryvasculature.

We have shown that the formed platelets from infused megakaryocytes are normalinsize,withoutasignificantnumberofmicroparticles(Figure28),andwithout notableactivation(Figure29A)orextracytoplasmiccleavageofsurfaceGPIb α(Figure

29BandC).Moreovertheseformedplateletscanbeactivelyincorporatedintogrowing thrombiFigure210.However,whythesederivedplateletshaveaslightlyshorterhalf lifethaninfused,isolatedplateletsisunclear.Weanticipatedthatplateletsderivedfrom megakaryocyteswouldbenewlygeneratedwithaprolonghalflife.

Whethertheshortenedhalflifehastodowiththeinvitroculturenecessaryto

grow the megakaryocytes is unknown. Previously, it had been suggested that

metalloproteinase ADAM17 is present in the media and affects platelet halflife (120).

Our studies showed no loss of at least one of the targeted surface marker by the

metalloproteinase and using the same inhibitors to block ADAM17 as previously

described did not alter the halflife of the platelets formed from infused large 44

megakaryocytes(Figure27).Analternativeexplanationfortheshortenedhalflifetothe

invitro culture conditions is that most of our studies were done with FLderived

megakaryocytesandfetalplateletshaveashortenedhalflifecomparedtoadultplatelets

(99).However, platelets derived from grown adult marrowmegakaryocytes also hada relatively short halflife than infused, donorderived platelets. There are insufficient numbersofprimarymegakaryocytesinisolatedadultmarrowtotestwhetherthesecells wouldhavealongerhalflife.

Ourdemonstrationthatinfusedmegakaryocytescanformasignificantnumberof

functionalplateletsmaybeofclinicalimportance.Invitrogenerationofalargenumber

of biologically responsive platelets has remained problematic. The bestpublished

outcomeforinvitroformedplateletsresultedwithlevelsofplateletsthatareatleasttwo

ordersofmagnitudelowercomparedtowhatwehavebeenabletoachievewithinfused

megakaryocytes.Ourstudiesopenthepossibilitythatifonecanestablishacelllinethat

canbeexpandedanddifferentiatedintomaturemegakaryocytesthenonemightbeable

toinfusethesemegakaryocytesasanondonordependentsourceofplatelets.Suchaline

had been described for mice, G1ME cells (121). These cells can be repeatedly derived

fromaGATA1deficientembryonicstemcelllineandthatuponrestorationofGATA1

goes on to form ~50% megakaryocytes (121). Moreover, modification of such lines to

ectopicallyexpressaproteinofinterestduringmegakaryopoiesismaybeusefulforthe

targeteddeliveryofsuchaproteintositesofvascularinjuryasdescribedbyusforthe

deliveryofcoagulationfactorVIIIforthetreatmentofhemophiliaAandurokinasefor

targeted fibrinolysis (84, 122). This additional benefit of generating platelets invivo

from infused modified megakaryocytes is supported by our data in Figure 211 that

shows that urokinase delivery to thrombi is a potential benefit of our approach using

mUK FLderived megakaryocytes that ectopically express urokinase and store this

45

proteinin αgranulesastheymature.

In summary, we have addressed whether infused megakaryocytes can release

platelets within the pulmonary bed and demonstrated thatbiologically active platelets

can be formed invivo with characteristics similar to those of normal platelets. The

process is vigorous, and enough platelets are formed so that platelet count can be boostedandhemostasiscanbeimprovedandmodifiedbytargeteddeliveryofectopic

proteins. Based on our estimate of 100–200 platelets generated per infused

megakaryocyte,thechallengeistogenerate10 9matureWTormodifiedmegakaryocytes

expressing and storing an ectopicprotein of interest to achieve a 10% rise in platelet

countinanaverage70kgpatient.

46

Table22.Supplementalmovielegend

Video 1. Incorporation of donor platelets in arteriole thrombi of h αIIb + recipientmouseafterinfusionofFLsmallcells,followinglaserinjury. Asin Figure210(C)withaninjurycreatednearthecenterofthevideoandbloodflowfrom toptobottomonthescreen.Theplateletsdepictedingreenweredetectedwithananti mouseCD41antibodylabeledwithAlexa 488 .Incorporationofplateletsfromsmallcells into the thrombi wasseen with a distinct population ofCD41 + cells recirculating and rarelyincorporatedintothegrowingthrombus. Video 2. Incorporation of donor platelets in arteriole thrombi of h αIIb + recipientmouseafterinfusionofFLlargecells,followinglaserinjury. Asin Figure210(B)withaninjurycreatednearthecenterofthevideoandbloodflowfrom toptobottomonthescreen.Theplateletsdepictedingreenweredetectedwithananti mouse CD41 antibody labeled with Alexa 488 . Incorporation of platelets from the large cellsintothegrowingthrombiwasdetectedsimilartoinfusionofWTplatelets. Video3.IncorporationofWTdonorplateletsinarteriolethrombiofh αIIb + recipient mouse following laser injury. As in Figure 210 (A) with an injury creatednearthecenterofthevideoandbloodflowfromtoptobottomonthescreen. TheplateletsdepictedingreenweredetectedwithanantimouseCD41antibodylabeled withAlexa 488 .IncorporationoftheWTinfusedplateletsintothegrowingthrombiwas clearlydetected.

47

Chapter3

Platelettargeted prourokinase as a novel

thromboprophylaxisfibrinolyticstrategy

3.1Abstract

Use of plasminogen activators (PAs) is restricted to lifethreatening thrombotic

conditions because high concentrations are required to diffuse into clots in order to

overcomePAinhibitorsandcompensateforrapidclearance,thatcanleadtobleeding.

We hypothesized that targeting proPAs to platelets would circumvent these obstacles

and preferentially lyse nascent, pathological clots that are actively recruiting platelets, whilesparingpreformedhemostaticclotsandalsoavoidingsystemicfibrinolysis.Totest

this concept, we produced a chimeric protein by fusing a scFv from an antihαIIb monoclonal antibody (moAb) 312.8 linked to a thrombin activatable uPAT, which we reasonedwouldbeactivatedpreferentiallyatsitesofactiveclotpropagation.AntiPLT scFv/uPATexpressedinDrosophilaS2insectcellsboundspecificallytohumanandto hαIIb + mouse platelets, but not to WT. AntiPLT scFv/uPAT bound specifically to

immobilized h αIIb β3 protein with a Kd of ~80 nM, and retained its zymogenic

propertiesuntilactivatedspecificallybythrombin.ThefibrinolyticactivityofantiPLT

scFv/uPAT and the nontargeted counterpart uPAT were analyzed in two mouse

+ modelsofvascularthrombosis.IntheFeCl 3carotidarteryinjurymodelh αIIb micewere

48

protectedfromformingocclusivethrombiforatleast10hrspostinjectionofantiPLT

scFv/uPAT whereas even fivefold higher concentrations of uPAT were effective for

only 25 min. In the cremaster arteriole injury model h αIIb + mice show a significant

reductioninbothfibrindepositionandplateletaccumulationevenafter5hrofantiPLT

scFv/uPAT infusion, which was not seen after infused uPAT or PBS. These studies

supportanothernovelapproachtothatinChapter2usingmodifiedmegakaryocytesfor

prophylactictargetingdrugdeliverybycombiningaprodrugthatrequiresactivationby

thrombinwithplateletdeliverytositesofincipientthromboticvascularocclusion.

3.2Background

Thrombosis is the major cause ofdeath and disability in the U.S.(80, 123). Occlusive

thrombi impairs blood flow to vital organs causing local oxygen deprivation, tissue

necrosisandorgandysfunction.PAsanddirectthrombininhibitorsareeffectiveacutely, but are associated with substantive risks of bleeding and are not formulated for

thromboprophylaxis. No agent preventsthrombosis in the majority of patients and all

increase the risk of bleeding, in large part because they act systemically. In theory, a

moreidealthromboprophylacticagentwouldhavethefollowingproperties:1)circulate

asaprodrug,2)targetincipientthrombi,whilesparinghemostaticclots,and3)remain

amenabletoregulation.

PAsarenotusedforprophylaxisduetotheirrapidclearanceandhemorrhagic

andneurotoxicsideeffects.AllexistingPAsaresmall,shortlived(<30min)molecules

capableofdiffusingintohemostaticclot,whichmaycausebleeding.PAscanalsodiffuse

into the CNS where they may cause collateral damage, and therefore are not 49

suited for use as prophylaxis in postsurgical or other patients at imminent risk of

thrombosis.WehypothesizedthatcouplingPAstoalongerlivedcarrierthatisconfined

tothebloodandhasanaturaltargetingmechanismtositesofthrombusformationwill blockPAdiffusionintohemostaticmuralclotsandtheCNS,limitingitsadverseeffects, while being retained in the blood, prolonging the intravascular effect, and thus permitting use as prophylaxis. We propose using a plateletdelivery strategy to target incipientorongoingpathologicalclotting.Todoso,wedevelopedarecombinantfusion protein composed of a scFv of a moAb (Figure 31) against the haIIb subunit of the platelet integrin receptor aIIbb3 (GPIIb/IIIa, CD41a) and a thrombinactivatable low molecularweightsinglechainurokinaseplasminogenactivator(lmwscuPA),whichhas beendesignateduPAT(Figure32).

3.3Materialandmethods

3.3.1AnalysisofantihαIIbmoAbs

Various antiplatelet moAbs were analyzed to check their binding specificity to WT,

hαIIb +miceandhumanplateletsbyflowcytometry(Figure35)(generouslyprovidedby

Dr.RichardAsterandDr.DanielBougiefromthemonoclonallaboratoryoftheBlood

Research Institute, Milwaukee, WI). The moAb (312.8) that we end up using in our

studieshasbeenpreviouslycharacterized(124).Murineplateletswereisolatedfromthe

inferior vena cava as described in“2.3.4 Isolation of Platelets and Megakaryocytes for

Infusion”. Human platelets were isolated from 10 ml of fresh blood, and platelet rich

plasma (PRP) separated after centrifugation at 200 g for 10 min at room temperature

(RT).Theplateletswerethenisolatedfrom PRPin thesamemannerasdescribefor 50

xx

Figure31.Finalconstructproposed. Theanticipatedfinalconstructisshownon

the right andwouldconsist of a singlechain version of uPA thatcontains a thrombin

cleavagesiteuPAT.Uponthrombinproteolysis,uPATwouldactivateplasminogento

plasminandleadtoclotlysisviafibrinolysis.TheuPATwillbefusedatitsNterminus

toanscFvderivedfromamoAb(shownonleft)thatbindsinaspecificfashiontothe

surfaceofhumanplateletsviathe αIIbreceptor

51

Figure 32. Molecular design of uPAT. Thrombininducible lmwscuPA was generatedbydeletingPhe 157 andLys 158 ,whichconvertstheplasminactivationsiteinto onethatiscleavebythrombinafterArg 156 .

52

murine platelets. Total washed platelets were counted on a Hemavet cell counter

(Triad Associates), resuspended in aliquots with a final concentration of 3x10 8/ml.

PlateletssuspensionswereincubatedwithcorrespondingmoAbs(0.1mg/ml)for1hrat

37ºC. Unbound moAb was washed with PBS prior to incubating with a secondary

polyclonalAb,FITCgoatantimouseIgG(BDBioscience)for30minandanalyzedby

flowcytometryusingaFACScan.

3.3.2PlateletaggregationofantihαIIbmoAbs

Plateletaggregationstudieswereperformedaspreviouslydescribed(98),Brieflyhuman

PRPwasadjustedto3X10 5platelets/ lwithplateletpoorplasma(PPP).SamplesofPRP were either untreated or incubated for 5 minutes at 37°C with 20 g/mL of moAb.

PlateletaggregationwasinducedbyaddingtoPRPadenosinediphosphate(ADP)at5

M,andlighttransmissionwasmeasuredovertimeinanaggregometer(KowaAG10E;

Kowa)whilestirring.

3.3.3ConstructionandexpressionofantiPLTscFv/uPAT

ConstructionoftheAntiPLTscFv/uPATwasperformedfollowingtechniquesusedfor fusingplasminogenactivatorswithserinerichlinkerpeptides,withsomemodifications

(85, 125, 126). Total RNA isolated from the moAbs hybridoma cell lines was reverse transcribed(RNeasy,Qiagen)usingSMART TM technology(Clontech)employingprimer

combinationsdescribedpreviously(127).Theheavyandlightchainvariablefragments were clone into TOPO vectors (Invitrogen) (Figure 33) and positive clones were

sequenced analyzed. The fragments were constructed using standard nestedPCR

methodology, in which the variable heavy (Vh) fragment was amplified with an

xxxxxxxxx

53

Figure 33. Depiction of the strategy to generate the antiPLT scFv. The schematic diagram depicts the strategies undertaken to produce the antiPLT scFv fragmentasdetailedintheMethodology. 54

Figure 34. Depiction of the strategy to generate the antiPLT scFv/uPAT.

Theschematicdiagramdepictsthestrategiesundertakentoproducethefinalconstruct, which was used to produce the fusionprotein antiPLT scFv/uPAT asdetailed in the

Methodology.

55

outerprimer(VhFWD5’tgcatgtgcatgccatggcagtcagggggaggc3’)andareverse

primerthatintroducedalinker(GGGGS) 3(128)attheendofthefragment(linkerrev5’ gattgctggagattgagtgagcacgctgccaccaccgccgctgccaccaccgccgctgccaccaccgccggg tgt cgt ttt ggc tga gga gac3’). The variable light (Vl) fragment was amplified with a complementaryprimerto“linkerrev”(linkerFWD5’gtctcctcagccaaaacgacacccggc ggtggtggcagcggcggtggtggcagcggcggtggtggcagcgtgctcactcaatctccagcaatc3’)and a downstream primer (Vl rev 5’ tcc ccg cgg agt tgg tgc agc atc3’). Joining of both fragments was completed by using the same outer primer “Vh FWD” that contains a restriction site for NcoI at the 5’ end, and the downstream primer “Vl rev” which

introducesarestrictionsitefor SpeI atthe3’endinordertocloneintoinsectexpression vectorpMT/BiP/V5HisB(Invitrogen)(Figure37A).TheantiPLTscFvPCRproducts were purified and digested with NcoI and SpeI restriction enzymes, (New England

BioLabs) ligatedandclonedintothe NcoI and SpeI sitesoftheinsectvector.Successful cloning was confirmed by restriction analysis of plasmid and by automated DNA sequencing(NAPCORE,TheChildren’sHospitalofPhiladelphia).Wethenestablisheda stableS2 drosophila cell line (Invitrogen) expressingthe antiPLT scFv(Figure37 C) andanalyzedtheproteinforbindingtoh αIIbfromtheinsectmediabyELISA(Figure3

8)andflowcytometry(Figure39).

Toproducethecompletechimericproteinconstruct,nestedPCRwasperformed

(Figure 34) to link the two fragments scFv and uPAT. The fragment for the scFv portionwasamplifiedwithprimer“VhFwd”andanewprimerthatintroducealinker

(SSSSG) 2 (128)downstreamofthescFv(linkerrev5’agtcttttggccacactgaaattttaagcc ggaagagctactacccgatgaggaagaagttggtgcagcatcagcccgttttatttcca3’).TheuPAT fragmentwasamplifiedwithacomplimentarylongprimertothe“linkerrev”(5’tggaaa taaaacgggctgatgctgcaccaacttcttcctcatcgggtagtagctcttccggcttaaaatttcagtgtggcc

56

aaaagact3’)andanouterprimer(uPAtrev5’ccaatgcattggctcgagtcatcacttgtcatc

gtcatccttgtaatcgat3’).

To join both fragments once again outer primer “Vh Fwd” which contain a

restrictionsitefor NcoI atthe5’endwasused,andthedownstreamprimer“uPATRev” whichintroducesarestrictionsitefor XhoI atthe3’endaswellasathreeflagtagwitha

stopsequence(Figure37B).Therestrictionsiteswereintroducedtocloneintoinsect

expressionvectorpMT/BiP/V5HisA.StabledrosophilacelllineexpressingtheantiPLT

scFv/uPAT was established for production of the protein. The proteins were purified

from cell media by affinity chromatography using an M2 (antiflag) affinity column

(Sigma) followed gel filtration chromatography on Sephacryl SHR100 column

(Amersham) to greater than 95% purity, which was confirmed by sodium dodecyl

sulfate–polyacrylamide gel electrophoresis (SDSPAGE) (Figure 310 A and B) with a yield approximately 3 to 5 mg/L medium. Proteins were concentrated in PBS to 2

mg/mL,separatedintoaliquotsandstoredat80°C.

3.3.4BiochemicalcharacterizationofantiPLTscFv/uPAT Characterization of the protein was performed in the same manner as previously

describedforotherscFvchimericproteins(126).Thesizeandhomogeneityofthefusion

proteinwasanalyzedusinga4%12%gradientSDSPAGEandWesternblotting(Figure

310 C). For Western blot analysis, the separated proteins were transferred to a

nitrocellulose membrane (Invitrogen) blocked with tris(hydroxymethyl)aminomethane

(Tris)buffered saline containing 10% nonfat milk powder and 0.1% Tween–20. The

fusionproteinwasdetectedusinga1:1000dilutionofahorseradishperoxidase(HRP)

conjugatedantiflagtagmoAb(Sigma).Theantigenantibodycomplexwasdetectedwith

ECLPlus(Amersham).

The intrinsic and thrombininduced activities of antiPLT scFv/uPAT were 57

measured using casein zymography. AntiPLT scFv/uPAT, (10 ng) intact or pre

incubatedwith20nMthrombin,weremixedwithnonreducingTrisglycineSDSsample bufferpriortozymography.Thesampleswereresolvedona7.5%gelcastwith10%non fat dry milk and 20 g/ml plasminogen to detect PA activity (129). The gels were re natured in Novex zymogram renaturating buffer (Invitrogen) and developed in Novex zymogramdevelopingbuffer(Invitrogen)perthemanufacturer.EDTA(5mM)(Sigma)

3.3.5BindingofantiPLTscFv/uPATtohαIIbβ3

BindingspecificityoftheantiPLTscFv/uPATtohαIIbb3wasanalyzedwithplatelets using flow cytometry techniques and the methodology described in “3.3.1 Analysis of antihαIIb monoclonal antibodies”. The specificity of the fusion protein was also analyzedwithplasticimmobilizedh αIIbh β3in2modes:bystandardELISA(Figure312

A)orwith 125 IlabeledantiPLTscFv/uPAT(Figure312B).H αIIbh β3protein(Enzyme

Research)wasimmobilizedonhighbindingplastic96wellplates(Corning)at1g/well.

Nonspecific binding was blocked by 5% PBS/BSA for 1 hr at 37°C then washed with

PBS/0.1%Tween20(PBS/Tween).ForstandardELISA,variousconcentrationsofanti

PLT scFv/uPAT, uPAT, or a nonspecific fusion protein antiGPA scFv/uPAT were incubatedfor1hrat37°C.AfterwashingwithPBS/Tween,anantiflagHRPconjugated moAbservedastheprimaryantibody.Theantigenantibodycomplexwasdetectedwith

3,3',5,5'tetramethylbenzidine(TMB)peroxidasesubstrate(Amersham)andabsorbance wasmeasuresat490nm.Forthesecondmethod,thefusionproteinwasradiolabeled with 125 INa(PerkinElmer)inIodinationtubes(ThermoScientific).Afterblockingwith

5% PBS/BSA and washing with PBS/Tween, various concentrations of the labeled chimericprotein(100l/well)wereincubatedwithh αIIbh β3orBSAcoatedwellsfor1hr at 37°C. Immobilized BSA served as a negative control. Nonbound material

58

wasremovedbywashingwithPBS/Tweenandresidualradioactivitywasmeasuredona

γcounter(PerkinElmer).

3.3.6InvitrofibrinolysisofantiPLTscFv/uPAT

Bovine fibrinogen (Sigma) was dissolved in PBS to a final concentration of 3 mg/ml.

Clotting was induced by addition of CaCl 2 and thrombin (Sigma) (20 mM and 0.2 units/mlfinalconcentrations,respectively)andtheresultingsolutionwasimmediately addedto24wellcellcultureplate(0.75ml/well)whereclotswereformedandallowedto maturate for 30 min. AntiPLT scFv/uPAT was activated by incubation with 20 nM bovinethrombin(Amersham)for1hrandserialdilutions(50l/well)wereaddedontop ofeachclotat37°C.Lyticzonesweremeasuredafter12hrincubation.

3.3.7EffectofantiPLTscFv/uPATinmodelsofvascularthrombosis

Studies were done on male h αIIb + mice as described in “2.3.7 Cremaster laser injury functionalstudies”. Thetiming,spatialandabsoluteincorporationoffibrinandplatelets intothegrowingthrombiwerefollowedusingaspecificantifibrinantibodyandananti speciesspecific αIIbAb,respectivelythatdonotinterferewithclotgrowth.

ForFeCl 3studies,weusedthestrategyasdescribedin“2.3.8FeCl 3carotidartery injury functional studies”. Briefly,1 mg/kgof antiPLT scFv/uPATor 1mg/kguPAT

+ was injected IV into h αIIb mice prior to FeCl 3induced carotid artery injury.

AdministrationofPBSservedasthenegativecontrol.Occlusionandtotalbloodflowwas monitoredbyDopplerultrasoundprobe.BothproteinsandPBSwereinjected10minsto

10hrspriortoarterialinjurytoestimatethetherapeuticwindowofthromboprophylaxis andbloodflowafterinjurywasmonitoredforover30min.

59

3.3.8FuturestudieswiththeantiPLTscFv/uPATfusionprotein

EffectofantiPLTscFv/uPATfusionproteinonplateletactivation. Toanalyzeif

antiPLT scFv/uPAT promotes activation of isolated platelets, flow cytometry will be

performedasdescribed(130)withminormodifications.Plateletsfromh αIIb +micewill be prepared and aliquot will be incubated with saturating concentrations of antiPLT

scFv/uPAT. Unbound protein will be removed by washing. As a positive control, a

subsetofintactplateletswillbestimulatedwiththeweakactivatorADP(25mmol/L)or buffer control. Fusion proteincoated platelets will be handled in the same manner.

Plateletsfromeachexperimentalgroupwillbefixedandwashed.Theplateletswillbe

stainedwithRphycoerythrin(PE)labeledPselectinantibody(mouseIgG,CLB),mouse

moAb PAC1 (Becton Dickinson), which recognizes only the activated form of mouse

αIIb β3,oranequivalentconcentrationofunspecificIgGandbindingwillbemeasured

usingaFACScan(131).

Effect of antiPLT scFv/uPAT on platelet aggregation . To further assess the

effect of the fusion proteins on platelet function, platelets from h αIIb + mice will be

incubated with saturating concentrations of antiPLT scFv/uPAT and the unbound

proteinwillberemovedbywashing.Aggregationinresponsetoaweakagonistwillbe

measuredusingaplateletaggregometerasdescribed(132).WTandunactivatedfusion

coatedh αIIb +murineplateletswillserveasnegativecontrols.

PharmacokineticsofantiPLTscFv/uPATvs.uPAT.AntiPLTscFv/uPATand

uPATwillbelabeledwith 125 Iandtraceramountswillbeinjectedviathejugularveinof

WTandh αIIb +mice.Bloodclearanceandmisdistributionoftheinjectedprotein10min,

60 min, 3, 6, 24, 48, and 72 hrs after infusion will be measured. The animals will be

sacrificedandmajororgans(lungs,liver,kidneys,spleen,heart,brain)willbeharvested

and perfused free of blood. PRP will be isolated and centrifuged at 10 4g to separate

60

plateletsfromPPP.Allbloodfractionsandorganswillbeanalyzedforradioactivityand

distributionofinjectedlabeledproteinswillbedetermined(%ofinjecteddoseand%of

injecteddose/gtissue).

Platelet counts will be measured to determine whether antiPLT scFv/uPAT

affects platelet lifespan. To verify that platelet halflife is not affected by the chimeric

protein in vivo, we will incubate h αIIb + mouse platelets with the fluorescent dense granule marker mepacrine (quinacrine) (133) and the fusion protein subsequently infusedintoh αIIb +mice.Survivaloftheinfusedplateletscanthenbedetectedbyflow

cytometryatdifferenttimepointsafterinfusion.

Wewillalsodeterminethedesorptionkineticof 125 IantiPLTscFv/uPATfrom

hαIIb +platelets.Washedplateletswillbeincubatedwith 125 IantiPLTscFv/uPATand

injectedintoWTorh αIIb +mice.Bloodandmajororganswillbecollectedasaboveand

thedistributionofresidualradioactivitywillbemeasuredovertime.Thehalflifeofthe

fusion protein loaded platelets will be determined using the Vybrant CFDA SE Cell

Tracerkit(MolecularProbes)aspreviouslydescribed(134).

Retention of antiPLT scFv/uPAT and uPAT fibrinolytic activity during

circulation in blood. We will follow the protocol described previously for a prototype

RBC/tPA conjugate (135, 136). Briefly, equimolar amounts of antiPLT scFv/uPAT,

uPATorPBSwillbeinjectedintravenously(IV)intoanesthetizedWTandh αIIb +mice.

At predesignated times determined above, aliquots of blood will be drawn in the

absence of anticoagulant, mixed rapidly with trace 125 Ifibrinogen and allowed to clot.

Clots willbeoverlaidwithPBSandincubatedat37°Cfor24hrsandthereleaseof 125 I willbemeasuredasabove.Theseexperimentswillallowustoestimatetherelationship betweenthedose,magnitudeanddurationoffibrinolyticactivityofantiPLTscFv/uPA

Tinthecirculation.

61

Depletion of fibrinogen by circulating antiPLT scFv/uPAT or uPAT. We will

followtheprotocoldescribedpreviouslyforaprototypeuPATfusionproteindirectedto

endothelium (85). Equimolar amounts of antiPLT scFv/uPAT, uPAT or PBS, in

amountsbasedontheblood fibrinolytic activity determined above, willbe injected IV

into WT and h αIIb + mice. At predesignated times based on the measured circulation

timeandbloodactivity,bloodwillbedrawnincitrateandPPP.Plasmafibrinogenwillbe

measuredbyELISAusingastandardcurvebasedonpurifiedmousefibrinogen.

BleedingafteradministrationofantiPLTscFv/uPATfusionprotein . Wewilluse

thetailrebleedingtimeasaninitialtestofhemostasisintreatedWTandh αIIb +mice

(137, 138). To evaluate hemorrhagic potential, mice will be anesthetized, equal doses

(determinedinexvivobloodclotlysisexperiments)ofantiPLTscFv/uPAToruPAT willbeinjected10minto72hrspriortoanalysis.Tailswillbeimmersedinawaterbath

at37°Cfor5min,a5mmterminalsegmentwillbeamputated,andthetimerequiredto

stopbleedingwillbedeterminedvisuallyandtheamountofreleasedhemoglobinwillbe

measured. After the initial bleeding has stopped, rebleeding will be evaluated by

immersingthetailin4mlof37°CPBScontaining14mmol/Ltrisodiumcitratefor1hr.

ThenumberofRBCandtheamountofhemoglobinreleasedinPBSwillbedetermined

to quantify blood loss (137, 138). To elucidate whether plateletbound antiPLT

scFv/uPATwillsparehemostaticclotsbetterthanuPAT,rebleedingwillbemeasured

as above, with the exception of injecting the proteins after spontaneous bleeding has

ceasedbeforeevaluatingrebleeding.

62

3.4Results

3.4.1BindinganalysisofantiplateletmoAbs

Binding of multiple antihαIIb moAbs (generously shared by Drs. Daniel Bougie and

Richard Aster, Blood Center of Wisconsin) was verify against WT, h αIIb + mice, and

human platelets and analyzed by flow cytometry. Many of the antibodies bound with

significant affinity to the surface of human platelets (Figure 35) and h αIIb + mice

platelets (not shown), but none to WT platelets (not shown). Two moAbs with high

affinitytohumanplateletswerechosenforfurtheranalysis(Abs184.2and312.8)(124).

Theantibodieswereanalyzedtoseeiftheyinterfereorenhanceplateletaggregation.As

itcanbeseeninFigure36,moAb184.2alteredaggregation,whereasmoAb312.8may

haveslightlyinhibitedaggregation.Bothwerefurtherstudiedunderstandingthatthere

levelofsurfaceaIIbb3receptorisdenseandatlowmoAbconcentrations,mostreceptors

maystillbeavailableforligandbinding.

3.4.2BindinganalysisofantiplateletscFv

ConstructionofthefusionproteinwasundertakenwithbothmoAbs(184.2and312.8) byisolatingRNAfromtheirhybridomacellsandperformingRTPCRtoformheavyand

lightvariablefragmentsbyanestedPCRstrategyasdepictedinFigure33.Theresultant

heavy and light chain variable fragments were assembled to form the antiPLT scFv

(Figure37A).ThepurifiedPCRproductwasclonedintoaninsectvectorandexpressed

inDrosophilaS2cellsandexpressionconfirmedbyimmunoblottingwithantiV5HRP

moAb (Figure 37 B). The overall construct was formed with the same PCR strategy

(Figure34)wherethescFvsegmentwasjoinedwithauPAT(Figure37C).Bindingof

x 63

Figure35.BindinganalysisofantihαIIbspecificmoAbstohumanplatelets.

Human platelets were incubated with 0.1 mg/ml of different antiplatelet moAb’s and

stainedwithanantimouseFITCconjugatedAbs.Meanfluorescenceof10 5 plateletswas measured. Only the two moAbs pursued further are noted. The binding to secondary antibody alone is shown as the grayfill graph. Details of the procedure are in the

Methodology.

64

Figure 36. Platelet aggregation analysis of antihαIIb specific moAbs. The twochosenmoAbswerestudiedfurtherusingplateletaggregationstudieswithhuman

PRPasdetailedintheMethodology.MonoclonalAbswereaddedat20g/mLandADP activationwasat5Matpointindicated(seearrows).

65

Figure37.GeneratingtheantiPLTscFv/uPATfusionconstruct .Thegeneral

constructstrategiesareshowninFigures33and34.(A)Agarosesizefractionationgel

oftheassembledscFvswithexpectedsizeof735basepairs.(B) Westernblotanalysisof nativeculturemediumfromS2cellstransfectedwithantiPLTscFvexpressionplasmid after induction with 0.5 mM CuSO 4. An HRPmoAb against the flag tag served as the detectingantibody,showingexpressionoftheexpectedscFvchimeraproteins(C)Same as (A), but for the scFvs assembled with a uPAT fragment into fulllength antiPLT scFV/uPATwithexpectedsizeof1640basepairs.

66

bothantiPLTscFv’stoimmobilizedh αIIbh β3proteinwasconfirmedbyELISA(Figure

38).Whenanalyzingbindingbyflowcytometry,antiPLTscFv312.8boundtohuman

andh αIIb +,butnotWT,mouseplatelets(Figure39A).AntiPLTscFv184.2boundto humanplatelets with less affinity (Figure 39A), anditdidnotbindtoh αIIb + orWT mouseplatelets(Figure39BandC).

3.4.3AnalysisofantiplateletscFv/uPAT

AfterestablishingstabledrosophilaS2celllinesexpressingantiPLTscFv/uPAT,both proteinswere purified fromcell media by affinity chromatographyusing an M2 (anti

FLAG) affinity column (Figure 310 A and B). To confirm that the fusion proteins retained the antigen recognition properties of the parental moAb and scFv, flow cytometry studies were performed. AntiPLT scFv/uPAT 312.8 bound to human platelets as well as h αIIb + but not WT mice platelets (Figure 311). The binding was similartowhatwasdemonstratedbyitsscFv.Interestingly,antiPLTscFv/uPAT184.2 appears to have lost its affinity since it did not recognize human or h αIIb + mouse platelets,incontrasttoitsscFv,likelyduetothechimericconstructdecreasingaffinityof thescFvforh αIIb(Figure311).Therefore,futurestudiesfocusedonthefusionprotein

312.8,andisreferredtobelowasantiPLTscFvuPAT.

TofurthershowthespecificityofantiPLTscFv/uPATweperformedtheexperimenton plasticimmobilizedh αIIb β3intwomodes:withstandardELISA(Figure312A)and 125 I labeled fusion protein (Figure 312 B). Both methods exhibited specific and dose dependent binding of antiPLT scFv/uPAT to the antigen with a Kd of ~80 nM.

Moreover,labelingwithiodinedidnotinfluencetheaffinityofthefusionprotein,which shouldsimplifyfuturepharmacokineticexperiments.

67

Figure38.ELISAanalysisofmoAbsandscFvinducedandnoninducedS2

media. ELISAof hαIIbh β3proteincoatedplateletswiththeoriginalindicatedmoAbor scFvS2insectcellmedia.Mean±1SDareshown.N=3,eachdoneinduplicate.

68

Figure39.CharacterizationofantiPLTscFv. (A)Human,(B)haIIb +mouseand

(C) and WT mouse platelets were incubated with 0.1 mg/ml of 312.8 antiPLT scFv

(red),moAb312.8(red)or184.2antiPLTscFv(green),andmoAb184.2(green)forflow cytometric studies of binding. Greyfilled in curve is secondary only binding. Mean fluorescenceof10 5 plateletswasmeasuredforeachcurve.

69

3.4.4FibrinolyticactivityofantiplateletscFv/uPATinvitro

To see if antiPLT scFv/uPAT has any PA activity in its native form, we performed

caseinzymography.WeconfirmedthatantiPLTscFv/uPATisexpressedasazymogen with negligible plasminogen conversion activity (Figure 313 A), which drastically increaseduponthrombinactivation.Thisresultappearstobesimilarthoughlesspotent than that seen with the nonfused thrombin activatable uPAT, which served as our positivecontrol.PurifiedantiPLTscFv/uPATmigratedasapredominantsinglebandat the predicted molecular weight of ~59 kDa and this was confirmed by Western blot

(Figure 313 B). Cleavage by thrombin generated two bands that under reducing conditionsresultsinaproteinfragmentsof~30kDa(Figure313B).

ToestimatethefibrinolyticactivityoftheantiPLTscFv/uPATuponthrombin conversionintoatwochainformcapableofactivatingplasminogen,fibrinplateanalysis wasperformed,whichrevealedstrong,dosedependentfibrinolyticactivityoftheanti

PLTscFv/uPAT,comparablewiththenonfuseduPAT(Figure313C).

3.4.5ThrombolyticefficacyofantiPLTscFv/uPATinmousemodels

ofvascularthrombosis

We tested whether antiPLT scFvuPAT thromboprophylaxis could effectively prevent occlusivethrombiusingtheFeCl 3modelofcarotidarterialinjuryinmiceaspreviously described(139).EquimolaramountsofantiPLTscFv/uPAT,unbounduPAT,orPBS

+ wereIVinjectedintoanesthetizedh αIIb micepriortoFeCl 3 carotidarteryinjury(98).

Inpreliminarydata(Figure314),itappearsthatadministrationofuPATorPBSdoes not prevent vascular occlusion after 5 min of infusion, while injection of antiPLT scFv/uPATwasprotectiveeven10hrspostinfusion.

Wealsousedthecremasterarteriole injury model, which allows for in situ 70

visualization

Figure310. CharacterizationoftheantiPLTscFV/uPATs. (A)and(B)SDS

PAGEgelsofelutedproteinsstainedwithGelCodebluestainreagent,and(C)Western blot of the antiPLT scFv/uPATs using an HRPmoAb against the flag tag as the

detectingantibody.

71

Figure 311. Flow cytometric analysis of haIIbb3 binding by antiPLT scFv/uPAT. (A) Human (A), (B) haIIb + mouse and (C) WT mouse platelets were incubated with 0.1 mg/ml of 312.8 antiPLT scFv/uPAT (red), moAb 312.8 (red) or

184.2 antiPLT scFv/uPAT (green), and moAb 184.2. Representative study of three separate flow cytometric analyses is shown. Mean fluorescence of 10 5 platelets was measured. 72

Figure 312. Binding of antiPLT scFv/uPAT to immobilized h αIIb β3. (A)

ELISAstudieswithbothhaIIbh β3inwellswiththevariousconcentrationsofantiPLT scFv/uPAT,uPAT,oraunrelatedfusionprotein,antiGPAscFv/uPATindicated.(B)

Binding of labeled antiPLT scFv/uPAT to immobilized h αIIbh β3. Varying concentrations of labeled antiPLT scFv/uPAT were incubated with h αIIbh β3 or BSA coatedwellsfor1hr.N=3,doneinduplicateeach.Mean±1SDofbindingisshown.

73

Figure 313. Enzymatic activity of antiPLT scFv/uPAT. The intrinsic and thrombininduced plasminogen activator activities of the antiPLT scFv/uPAT were analyzed using (A) casein zymography and (B) Western blot with a mouse anti lmw scuPA antibody (American Diagnostica) as primary antibody. (C) Fibrin plate analysis revealed a strong anddose dependent fibrinolytic activity of the antiPLT scFv/uPAT similartouPAT.

74

Figure 314. Prophylactic thrombolysis by antiPLT scFV/uPAT in a FeCl 3 carotidarteryinjurymodel. Indicatedproductswereinfusedattimezeroandthena

FeCl 3 injury followed by a 30min measurement of carotid artery blood flow was monitoredattheindicatedtimepoints.Mean±1SDareshown.N=3.

75

visualizationofthrombusdevelopmentinrealtimetotestourfusionprotein.Inthese

studies, h αIIb + male mice were IV infused with equimolar amounts of antiPLT

scFv/uPAT,unbounduPATorPBS5hrsbeforeinducing injury. Mice infused with

either PBS (Figure 315 A) or uPAT (Figure 315 B) showed similar fibrin deposition with only a small difference in platelet accumulation. Mice infused with antiPLT scFv/uPATfusionprotein(Figure315C)showedasignificantreductioninbothfibrin deposition and platelet accumulation. These preliminary studies support our fusion proteinasbeingbothaneffectiveandlonglastingthromboprophylacticagent.

3.5Discussion

Thromboticarterialocclusionistheproximatecauseofmyocardialinfarctionandstroke.

Occlusive thrombi impair blood flow to vital organs causing local oxygen deprivation, tissue necrosis andorgan dysfunction. CurrentPA therapy is not used forprophylaxis due to their rapid clearance, (<30 min) risk of bleeding and neurotoxic side effects.

NewlydesignedmutantPAswithenhancedpotencyandfewersideeffectsmayimprove this outcome (140145), but their selective diffusion into occlusive clots has not been testedandnonehasbeendesignedforprophylacticuse. Anidealthromboprophylactic agentneedstohaveaprolongedlifespanintheblood,circulateasaprodrug,andtarget incipientthrombiwhilesparinghemostaticclots.

RecentstudieshavetargetedPAstoredbloodscells(RBC)byreinfusingexvivo modifiedcells(139)andbyIVinjectionofPAsfusedtoeitherFab’orscFvfromantiRBC

(126, 146). These studies showed that the fused proteins provided prolonged thromboprophylaxis against lethal stroke and pulmonary embolism with less re 76

xxxxxxx

Figure 315. Platelet and fibrin accumulation in laserinduced arteriole

injuries in treated haIIb + mice. Analysis of in situ laser injury in cremaster arterioles5hrsposttreatmentwith(A)PBS,(B)uPAT,and(C)antiPLTscFvuPAT.

Averageplateletaccumulation(green)andfibrinclotaccumulation(red)areshownover a3minwindowafteralaserinjury.N=2miceperstudywith10injuriespermouse.

77

bleeding after tail amputation (125, 146) or intracerebral hemorrhage after traumatic braininjury(147).ButRBCsarenotspecificallytargetedtogrowingthrombi,especially arterialthrombi.Platelets,ontheotherhand,becomeincorporatedintoincipientclots, which makes them ideal for a thromboprophylaxis design. Our group has shown that

uPAectopicallyexpressedinplateletsusingatransgenicapproachissafeandeffectivein

preventing arterial and venous thrombosis and that transfusion of 5% of the platelet

mass protects wild type mice from clotting without causing systemic fibrinolysis or

spontaneousbleedingasidefrompostpartumhemorrhage(84).

WeusedasimilarstrategyastheRBCPAstudies,utilizingplateletstodeliverthe

thrombinactivatable mutant uPAT to newly formed clots. We hypothesize that this

approachwill;1)increasethehalflifeofuPATincirculation,2)significantlyincrease

deliveryofuPATtositesofincipientthrombusformation,3)limitsystemicsideeffects

duetolocalactivationofuPATbythrombin,and4)bypassmaturehemostaticclotsnot

activelyrecruitingplateletswithlessthrombinpresent.

We developed a recombinant fusion protein composed of a scFv from a moAb

(Figure 31) against theh αIIbsubunit of theplatelet integrinreceptor αIIb β3fused to

thrombinactivatable uPAT (Figure 32). The new zymogen, antiPLT scFv/uPAT,

4 boundspecificallytohumanplatelets,withaB max of~2x10 moleculespercell,assuming

approximately80,000GPIIb/IIIacomplexesonthesurfaceofeachplatelet(90).Anti

PLTscFv/uPATalsoboundtoh αIIb +,butnotWT,mouseplatelets(Figures311and3

12).Thefusionproteinexhibitedpronouncedactivity,butonlyuponthrombinactivation

andconversionofuPATintoitsactiveform(Figure313).Totestthromboprophylaxis, weusedtwomousemodelsofacutethromboticocclusioninresponsetovascularinjury.

IntheFeCl 3carotidarteryinjurymodel,itappearsthatuPATorPBSdoesnotprevent vascularocclusion5minafteradministration,whileantiPLTscFv/uPATwasprotective

78

even after 10 hrs post infusion (Figure 314). When analyzing h αIIb + mice with the cremasterarterioleinjurymodel5hrsafteradministeringtheantiPLTscFv/uPAT,we sawasignificantreductioninbothfibrindepositionandplateletaccumulationcompared tomicethathadbeentreatedwithuPATorPBS(Figure315).

Thesepreliminaryresultsfromthemousemodelsofvascularinjuryconcurwith ourexpectationthattheantiPLTscFv/uPATbindsrapidlytothesurfaceofplateletsin hαIIb + mice, while uPAT is rapidly eliminated from circulation through hepatic

clearance.Furtherstudiesshouldprovideuswithabetterpictureasitrelatestoplatelet

survival,biodistributionoftheproteininthelungs,liverorspleen,andthedurationof

fibrinolyticactivity.Italsoremainstobedeterminedwhethertheplateletboundfusion

proteininducesbleedingor,conceivably,rebleeding,atdosesthatpreventclotbuildup.

Weproposeapotentiallyclinicallyfeasiblemeanstotargetaprodrugusingthe

natural procoagulant plateletthrombin pathway to induce fibrinolysis and reduce the

need for higher dosage. If effective, the design of the fusion protein and targeting

strategy allows for adaptation to other prodrugs with diverse activities (anti

inflammatory, antioxidant) that can be targeted to platelets or other cell types, (e.g.,

,monocytes)thattargettositesofinjuryinotherdisorders.

79

Chapter4

Summary,andfuturedirections

PAsassistinmaintainingbloodfluiditybyactivatingplasmintodegradefibrinclots(62,

148, 149), but they have limited clinical utility due to their rapid clearance and problematic side effects, especially untoward bleeding. Their clinical limitations make

PAsunsuitableforprophylactictherapy,whichmerittheneedfornewerdrugsthatare safer and better suited for effective fibrinolysis. In theory, a more ideal thromboprophylactic agent should have the following properties: 1) has a prolonged lifespan in the blood, 2) circulates as a prodrug, 3) targets incipient thrombi, while sparinghemostaticclots,and4)remainsamenabletoregulation.Plateletsprovidethe firstlineofdefenseintheprocessofhemostasis,bylocalizingandsustainingtheblood lossandadheringtothesiteofinjury.Theirnaturaltargetingfunctionmakestheman ideal candidate for therapeutic delivery of bioactive agents. We concentrated on two distinct strategies that in proof of principle studies are promising for effective and targetedplateletdirectedthrombolytictherapy.

Inourfirstapproach,wewereinterestedintargetdeliveryofuPAtotheinjured sitebyexpressingtheproteinathighlevelsindevelopingmegakaryocytessothatitis stored in platelet alpha granules and released upon activation. These findings were previouslydemonstratedinourlaboratorywithtransgenicmicethatectopicallyexpress uPAwithinplatelets(84).TheuPAloadedplateletsshowedthattheycouldselectively lyseemboliwithoutinducingbleeding,systemicdecreaseinfibrinogenlevelsorincrease

80

in Ddimers. Expression of uPA within platelets also is the underlying mechanism in

Quebec platelet disorder, wherein patients have mild bleeding (105); supporting the

contention that platelets laden with uPA may be an effective PA with little systemic bleeding. These patients show significant posttraumatic bleeding, but this is

controllable with tranexamic acid (150153), suggesting a therapeutic strategy to treat

untowardbleedingfromuPAladenedplateletsasaPAstrategy.

Wewereinterestedintranslatingthesefindingsintoclinicalusebeginningwith exvivo generated megakaryocytes and platelets for thrombolytic targeted therapy. No one had successfully produced sufficient exvivo generated platelets before and it is possible that given the plastic surfaces and slow release of ex vivo generated platelets from megakaryocytes that we would have difficulty generating large numbers of non activated platelets. Because of older literature suggesting that circulating megakaryocytesmightbecomelodgedinthelungsandreleaseplateletsthereafter(35,

36, 39), we tested whether infusing exvivo generated megakaryocytes directly would leadtoreleasedplateletswithinthelungs.Wefoundthatwecanachieveasignificant number of donorderived platelets from these infused megakaryocytes in a murine modelwithadelayof390minutes.Whiletheseplateletshadaslightlyshortenedhalf lifecomparedtodonorderivedplatelets,theyhadmanyfeaturesofnormalplateletsand werefunctionalintwoinvivoassays.Inaradiationinducedthrombocytopeniamodel, we showed that these platelets can boost the platelet count as well as donorderived plateletsandinthissystem,mayevenlastbetter.Whetherthesemegakaryocytederived plateletsenhancedpostradiationsurvivalhasyettobetested.IntheFeCl 3carotidartery model,wenotonlytestedplateletefficacy,butshowedthatmegakaryocytesderivedfrom transgenic mice with plateletuPA expression, when infused into the recipient mouse completelypreventedthrombusdevelopment.Wetakethisasthefirststepinaproofof

81

principlethatourapproachofinfusingmodifiedmegakaryocytesmaybeamodelforthe

targeted delivery of a PAs to a site of untoward thrombosis and may be useful for

targetedthromboprophylaxis.

Inoursecondapproach,weextendedastrategydevelopedbyourcolleaguesof

endothelial and RBCs targeting fused to a thrombin activated proenzyme uPAT (85,

126) to platelets. They had shown targeted efficient and safe thromboprophylaxis in

several clinically relevant models including after pulmonary injury.Weextended their

strategy by using a plateletdelivery system with uPAT that is activated and released from the antibody by thrombin generated at the site of active clotting. Since platelets targetnewthrombibetterthanRBCs,webelievethattheremaybeadvantagesforthe bindingofuPATtotheplateletsurfaceovertheredcellsurface.OurfusionuPATthat

hadanscFvderivedfromanantihaIIbmoAbgenerouslyprovidedbymycolleaguesDrs.

RichardAsterandDanielBougieattheBloodCenterinWisconsin,boundspecificallyto

humanplateletsthroughits αIIbb3receptor.ThisfusionuPATalsoboundtotransgenic

micethathadchimerichaIIbmb3aretheirplatelets,butnottoWTplatelets.Thefusion

protein exhibited pronounced activity, but only upon thrombin activation and

conversion of uPAT into its active form. In preliminary thromboprophylactic

experimentsusingamousemodelofvascularinjury,itappearsthatthefusionprotein waslonglastingandpreventedthrombusdevelopmentforatleast10hrspostinfusion

intheFeCl 3carotidarteryinjurymodelwithoutobviousbleedingcomplications,while

uPAT or PBS did not prevent vascular occlusion 5 min after administration. In

analyzingh αIIb + micewiththecremasterarterioleinjurymodel5hrsafteradministering

ofthefusionprotein,wealsosawasignificantreductioninbothfibrindepositionand

plateletaccumulationcomparedtomicethathadbeentreatedwithuPATorPBS.These

dataarepreliminary,andneedtoberepeated.

82

Forthefuture,thestudiespresentedhereserveasproofofprincipleoftwonovel therapeuticapproachesforthedeliveryofuPAespeciallyforthromboprophylaxis.Both strategies include using platelets to target uPA to incipient thrombi and hopefully to avoid more mature thrombi and enhance efficacy and selectivity of the PA. For the megakaryocytederivedplateletstrategyourultimategoalwouldbetoestablishaself replicatinghumancelllinethatuponappropriatestimulusterminallydifferentiatesinto megakaryocytesexpressinguPA.Uponinfusionofthesecells,weanticipateefficacious release of platelets from these cells. For our second approach involving antiPLT scFv/uPAT, safety and pharmacokinetics and pharmacodynamic studies need to be completedinmice.APhaseIsafetytrialwouldbeneededinasettingassociatedwitha highriskofthrombosisthatmaybenefitfromthromboprophylaxis.Thismaybepatients undergoingstentplacementorasubgroupofsuchpatientsthatareathigherriskofsuch thrombi, like in patients with multivessel coronary artery disease with a higher prevalenceofcoronaryriskfactorswhencomparedtopatientswithout(154).

Ifeitherapproachprovestobeclinicallyuseful,thestrategiesdevelopedherefor platelettargetedtherapymaybeextendedtoothersystemssuchasthedeliveryofagents that are antiinflammatory, like thrombomodulin either stored within platelets or a recombinantthrombomodulinproteindesignedbyour colleague,BiSenDing(155)at the University of Pennsylvania with a thrombin cleavage site, transforming thrombin intoanantiinflammatorymolecule.Ourapproachcanalsobeadaptedtoproduceanti angiogenicagentsbydesigningplateletsstoredwithnegativeangiogenicregulatorssuch asthrombospondin1,endostatin,andangiostatintoinhibittumorprogression.Wecan also target the tumor with scFvs with a cleavage site for cathepsinB, which is over expressedinmanytumorcellsandtumorendothelialcells.Themodulardesignusedin ourscFvcanbeappliedtoothercellsthatcanbetargetedandareessentialintreating

83

othermalignanciessuchasinfectiousdisease.Forexampleneutrophilsandmonocytes canbetargetedviathemyeloidFc αRI(CD89)receptorwhichhasbeenidentifiedasan effectivetargetmolecule(156),andisexpressedexclusivelyinmyeloidcells.

84

References

1. Mackman, N. 2008. Triggers, targets and treatments for thrombosis. Nature 451:914918. 2. Cines, D.B., Pollak, E.S., Buck, C.A., Loscalzo, J., Zimmerman, G.A., McEver, R.P.,Pober,J.S.,Wick,T.M.,Konkle,B.A.,Schwartz,B.S.,etal.1998.Endothelial cells in physiology and in the pathophysiology of vascular disorders. Blood 91:35273561. 3. Francois, P., Verstraeten, L., and Dinant, J.P. 1989. [The physiology of hemostasis:plasmaandtissuefactorsincoagulationandfibrinolysis]. J Pharm Belg 44:308314. 4. Tocantins,L.M.1948.Historicalnotesonbloodplatelets. Blood 3:10731082. 5. Bizzozero, G. 1882.Ueber einer neuen formbestandtheil desblutes unddessen rollebeiderthromboseundderblutgerinnung. Virchows Arch fur pathol Anat und Physiol 90:261332. 6. Bizzozero, J. 1882. Über einen neuen Formbestandteil des Blutes und dessen RollebeiderThromboseundderBlutgerinnung. Virchow's Arch Path Anat Klin Med 90:261–332. 7. Howell,W.1890.Observationsupontheoccurrence,structure,andfunctionof thegiantcellsofthemarrow. J Morphol 4:117130. 8. Wright,J.H.1906.TheOriginandNatureoftheBloodPlates. Boston Med Surg J 154:643645. 9. Pang, L.,Weiss, M.J., and Poncz, M. 2005. Megakaryocyte biology and related disorders. J Clin Invest 115:33323338. 10. Golde,D.W.1991.Thestemcell. Sci Am 265:8693. 11. Ogawa, M. 1993. Differentiation and proliferation of hematopoietic stem cells. Blood 81:28442853. 12. Keller,G.,Lacaud,G.,andRobertson,S.1999.Developmentofthehematopoietic systeminthemouse. Exp Hematol 27:777787. 13. Vainchenker, W., Bouguet, J., Guichard, J., and BretonGorius, J. 1979. Megakaryocyte colony formation from human bone marrow precursors. Blood 54:940945. 14. Metcalf,D.,MacDonald,H.R.,Odartchenko,N.,andSordat,B.1975.Growthof mousemegakaryocytecoloniesinvitro. Proc Natl Acad Sci U S A 72:17441748. 15. Kaushansky,K.2008.Historicalreview:megakaryopoiesisandthrombopoiesis. Blood 111:981986. 16. Nakorn,T.N.,Miyamoto,T.,andWeissman,I.L.2003.Characterizationofmouse clonogenicmegakaryocyteprogenitors. Proc Natl Acad Sci U S A 100:205210. 17. Vinci, G., Tabilio, A., Deschamps, J.F., Van Haeke, D., Henri, A., Guichard, J., Tetteroo, P., Lansdorp, P.M., Hercend, T., Vainchenker, W., et al. 1984. Immunological study of in vitro maturation of human megakaryocytes. Br J Haematol 56:589605. 18. Long, M.W., and Williams, N. 1981. Immature Megakaryocytes in the Mouse: Morphology and quantitation by acetylcholinesterase staining. Blood 58:1032 1039. 19. Therman,E.,Sarto,G.E.,andStubblefield,P.A.1983.Endomitosis:areappraisal. Hum Genet 63:1318. 85

20. Hogge,D.,Fanning,S.,Bockhold,K.,Petzer,A.,Lambie,K.,Lansdorp,P.,Eaves, A., and Eaves, C. 1997. Quantitation and characterization of human megakaryocyte colonyforming cells using a standardized serumfree agarose assay. Br J Haematol 96:790800. 21. Kaushansky, K. 1995. Thrombopoietin: the primary regulator of platelet production. Blood 86:419431. 22. Flanagan,J.G.,andLeder,P.1990.Thekitligand:acellsurfacemoleculealtered insteelmutantfibroblasts. Cell 63:185194. 23. Briddell,R.A.,Bruno,E.,Cooper,R.J.,Brandt,J.E.,andHoffman,R.1991.Effect ofckitligandoninvitrohumanmegakaryocytopoiesis. Blood 78:28542859. 24. Battinelli,E.M.,Hartwig,J.H.,andItaliano,J.E.,Jr.2007.Deliveringnewinsight into the biology of megakaryopoiesis and thrombopoiesis. Curr Opin Hematol 14:419426. 25. Kaushansky,K.2002.Thrombopoietin:fromtheorytoreality. Int J Hematol 76 Suppl1:343345. 26. Kaushansky, K. 2002. Mpl and the hematopoietic stem cell. Leukemia 16:738 739. 27. Rock,G.,andTittley,P.1990.Acomparisonofresultsobtainedbytwodifferent chromium51methodsofdeterminingplateletsurvivalandrecovery. Transfusion 30:407410. 28. Stenberg, P.E., and Levin, J. 1989. Mechanisms of platelet production. Blood Cells 15:2347. 29. Djaldetti,M.,Fishman,P.,Bessler,H.,andNotti,I.1979.SEMobservationson the mechanism of platelet release from megakaryocytes. Thromb Haemost 42:611620. 30. Ihzumi, T., Hattori, A., Sanada, M., and Muto, M. 1977. Megakaryocyte and plateletformation:ascanningelectronmicroscopestudyinmousespleen. Arch Histol Jpn 40:305320. 31. Shaklai, M., and Tavassoli, M. 1978. Demarcation membrane system in rat megakaryocyte and the mechanism of platelet formation: a membrane reorganizationprocess. J Ultrastruct Res 62:270285. 32. Becker,R.P.,andDeBruyn,P.P.1976.Thetransmuralpassageofbloodcellsinto myeloid sinusoids and the entry of platelets into the sinusoidal circulation; a scanningelectronmicroscopicinvestigation. Am J Anat 145:183205. 33. Italiano, J.E., Jr., Lecine, P., Shivdasani, R.A., and Hartwig, J.H. 1999. Blood plateletsareassembledprincipallyattheendsofproplateletprocessesproduced bydifferentiatedmegakaryocytes. J Cell Biol 147:12991312. 34. Junt,T.,Schulze,H.,Chen,Z.,Massberg,S., Goerge, T., Krueger, A., Wagner, D.D., Graf, T., Italiano, J.E., Jr., Shivdasani, R.A., et al. 2007. Dynamic visualizationofthrombopoiesiswithinbonemarrow. Science 317:17671770. 35. Kaufman, R.M., Airo, R., Pollack, S., and Crosby, W.H. 1965. Circulating megakaryocytesandplateletreleaseinthelung. Blood 26:720731. 36. Pedersen,N.T.1978.Occurrenceofmegakaryocytesinvariousvesselsandtheir retentioninthepulmonarycapillariesinman. Scand J Haematol 21:369375. 37. Tavassoli, M. 1980. Megakaryocyteplatelet axis and the process of platelet formationandrelease. Blood 55:537545. 38. TinggaardPedersen,N.,andCohn,J.1981.Intactmegakaryocytesinthevenous bloodasamarkerforthrombopoiesis. Scand J Haematol 27:5763. 39. ZuckerFranklin, D., and Philipp, C.S. 2000. Platelet production in the pulmonarycapillarybed:newultrastructuralevidenceforanoldconcept. Am J Pathol 157:6974. 86

40. Sere, K.M., and Hackeng, T.M. 2003. Basic mechanisms of hemostasis. Semin Vasc Med 3:312. 41. Hirsh, J., Anand, S.S., Halperin, J.L., and Fuster, V. 2001. AHA Scientific Statement:Guidetoanticoagulanttherapy:heparin:astatementforhealthcare professionals from the American Heart Association. Arterioscler Thromb Vasc Biol 21:E99. 42. Schafer, A.I., Zavoico, G.B., Loscalzo, J., and Maas, A.K. 1987. Synergistic inhibitionofplateletactivationbyplasminandprostaglandinI2. Blood 69:1504 1507. 43. Radomski, M.W., Palmer, R.M., and Moncada, S. 1991. Modulation of platelet aggregationbyanLargininenitricoxidepathway.Trends Pharmacol Sci 12:87 88. 44. Marcus,A.J.,Broekman,M.J.,Drosopoulos,J.H.,Islam,N.,Alyonycheva,T.N., Safier,L.B.,Hajjar,K.A.,Posnett,D.N.,Schoenborn,M.A.,Schooley,K.A.,etal. 1997. The endothelial cell ectoADPase responsible for inhibition of platelet functionisCD39. J Clin Invest 99:13511360. 45. Brass,L.F.2003.Thrombinandplateletactivation. Chest 124:18S25S. 46. Frenette,P.S.,Johnson,R.C.,Hynes,R.O.,andWagner,D.D.1995.Plateletsroll on stimulated endothelium in vivo: an interaction mediated by endothelial P selectin. Proc Natl Acad Sci U S A 92:74507454. 47. Frenette,P.S.,Moyna,C.,Hartwell,D.W.,Lowe,J.B.,Hynes,R.O.,andWagner, D.D. 1998. Plateletendothelial interactions in inflamed mesenteric venules. Blood 91:13181324. 48. DeMarco,L.,Girolami,A.,Russell,S.,andRuggeri, Z.M. 1985. Interaction of asialovonWillebrandfactorwithglycoproteinIbinducesfibrinogenbindingto the glycoprotein IIb/IIIa complex and mediates platelet aggregation. J Clin Invest 75:11981203. 49. Farndale,R.W.,Sixma,J.J.,Barnes,M.J.,anddeGroot,P.G.2004.Theroleof collageninthrombosisandhemostasis. J Thromb Haemost 2:561573. 50. Bevilacqua,M.P.,andGimbrone,M.A.,Jr.1987.Inducibleendothelialfunctions ininflammationandcoagulation. Semin Thromb Hemost 13:425433. 51. Coughlin,S.R.2005.Proteaseactivatedreceptorsinhemostasis,thrombosisand vascularbiology. J Thromb Haemost 3:18001814. 52. Wagner,D.D.,andBurger,P.C.2003.Plateletsininflammationandthrombosis. Arterioscler Thromb Vasc Biol 23:21312137. 53. Phillips, D.R., NannizziAlaimo, L., and Prasad, K.S. 2001. Beta3 tyrosine phosphorylation in alphaIIbbeta3 (platelet membrane GP IIbIIIa) outsidein integrinsignaling. Thromb Haemost 86:246258. 54. Pasche,B.,andLoscalzo,J.1991.PlateletsandFibrinolysis. Platelets 2:125134. 55. Bass, R., and Ellis, V. 2002. Cellular mechanisms regulating nonhaemostatic plasmingeneration. Biochem Soc Trans 30:189194. 56. Booth, N.A., Simpson, A.J., Croll, A., Bennett, B., and MacGregor, I.R. 1988. Plasminogenactivatorinhibitor(PAI1)inplasmaandplatelets. Br J Haematol 70:327333. 57. Booth,N.A.,Robbie,L.A.,Croll,A.M.,andBennett,B.1992.Lysisofplateletrich thrombi:theroleofPAI1. Ann N Y Acad Sci 667:7080. 58. Kruithof, E.K., Nicolosa, G., and Bachmann, F. 1987. Plasminogen activator inhibitor1:developmentofaradioimmunoassayandobservationsonitsplasma concentration during venous occlusion and after platelet aggregation. Blood 70:16451653. 87

59. Raum,D.,Marcus,D.,Alper,C.A.,Levey,R.,Taylor,P.D.,andStarzl,T.E.1980. Synthesisofhumanplasminogenbytheliver. Science 208:10361037. 60. Dobrovolsky,A.B.,andTitaeva,E.V.2002.Thefibrinolysissystem:regulationof activityandphysiologicfunctionsofitsmaincomponents. Biochemistry (Mosc) 67:99108. 61. Levin,E.G.,andLoskutoff,D.J.1982.Culturedbovineendothelialcellsproduce bothurokinaseandtissuetypeplasminogenactivators. J Cell Biol 94:631636. 62. Collen, D., and Lijnen, H.R. 2005. Thrombolytic agents. Thromb Haemost 93:627630. 63. Hoylaerts, M., Rijken, D.C., Lijnen, H.R., and Collen, D. 1982. Kinetics of the activationofplasminogenbyhumantissueplasminogenactivator.Roleoffibrin. J Biol Chem 257:29122919. 64. Tkachuk,V.,Stepanova,V.,Little,P.J.,andBobik,A.1996.Regulationandroleof urokinase plasminogen activator in vascular remodelling. Clin Exp Pharmacol Physiol 23:759765. 65. Clowes,A.W.,Clowes,M.M.,Au,Y.P.,Reidy,M.A.,andBelin,D.1990.Smooth musclecellsexpressurokinaseduringmitogenesisandtissuetypeplasminogen activatorduringmigrationininjuredratcarotidartery. Circ Res 67:6167. 66. Eaton,D.L.,Scott,R.W.,andBaker,J.B.1984.Purificationofhumanfibroblast urokinase proenzyme and analysis of its regulation by proteases and protease nexin. J Biol Chem 259:62416247. 67. Bacharach, E., Itin, A., and Keshet, E. 1992. In vivo patterns of expression of urokinase and its inhibitor PAI1 suggest a concerted role in regulating physiologicalangiogenesis. Proc Natl Acad Sci U S A 89:1068610690. 68. Holmes,W.E.,Pennica,D.,Blaber,M.,Rey,M.W.,Guenzler,W.A.,Steffens,G. L., and Heyneker, H. L. 1985. Cloning an expression of the gene for pro urokinaseinescherichiacoli. Nature Biotechnology 3:923929. 69. Appella,E.,Robinson,E.A.,Ullrich,S.J.,Stoppelli,M.P.,Corti,A.,Cassani,G., and Blasi, F. 1987. The receptorbinding sequence of urokinase. A biological functionforthegrowthfactormoduleofproteases. J Biol Chem 262:44374440. 70. Mimuro, J., Kaneko, M., Murakami, T., Matsuda, M., and Sakata, Y. 1992. Reversible interactions between plasminogen activators and plasminogen activatorinhibitor1. Biochim Biophys Acta 1160:325334. 71. Kobayashi,H.,Gotoh,J.,Hirashima,Y.,Fujie,M.,Sugino,D.,andTerao,T.1995. Inhibitory effect of a conjugate between human urokinase and urinary trypsin inhibitorontumorcellinvasioninvitro. J Biol Chem 270:83618366. 72. Ugwu,F.,VanHoef,B.,Bini,A.,Collen,D.,andLijnen,H.R.1998.Proteolytic cleavage of urokinasetype plasminogen activator by stromelysin1 (MMP3). Biochemistry 37:72317236. 73. Lijnen, H.R., Van Hoef, B., Nelles, L., and Collen, D. 1990. Plasminogen activation with singlechain urokinasetype plasminogen activator (scuPA). Studieswithactivesitemutagenizedplasminogen(Ser740Ala)andplasmin resistantscuPA(Lys158Glu). J Biol Chem 265:52325236. 74. Collen, D. 1996. Fibrinselective thrombolytic therapy for acute myocardial infarction. Circulation 93:857865. 75. Gruntzig,A.R.,Senning,A.,andSiegenthaler,W.E.1979.Nonoperativedilatation ofcoronaryarterystenosis:percutaneoustransluminalcoronaryangioplasty. N Engl J Med 301:6168. 76. Verstraete,M.2000.Thirdgenerationthrombolyticdrugs. Am J Med 109:5258. 88

77. Collen, D., De Cock, F., and Stassen, J.M. 1993. Comparative immunogenicity andthrombolyticpropertiestowardarterialandvenousthrombiofstreptokinase andrecombinantstaphylokinaseinbaboons. Circulation 87:9961006. 78. Longstaff, C., and Thelwell, C. 2005. Understanding the enzymology of fibrinolysisandimprovingthrombolytictherapy. FEBS Lett 579:33033309. 79. VandeWerf,F.1993.Newthrombolyticstrategies. Aust N Z J Med 23:763765. 80. Jackson, M.R., and Clagett, G.P. 1998. Antithrombotic therapy in peripheral arterialocclusivedisease. Chest 114:666S682S. 81. Augustinos,P.,andOuriel,K.2004.Invasiveapproachestotreatmentofvenous thromboembolism. Circulation 110:I2734. 82. Hass, W.K., Easton, J.D., Adams, H.P., Jr., PrysePhillips, W., Molony, B.A., Anderson, S., and Kamm, B. 1989. A randomized trial comparing ticlopidine hydrochloride with aspirin for the prevention of stroke in highrisk patients. TiclopidineAspirinStrokeStudyGroup. N Engl J Med 321:501507. 83. Yach,D.,Hawkes,C.,Gould,C.L.,andHofman,K.J.2004.Theglobalburdenof chronic diseases: overcoming impediments to prevention and control. JAMA 291:26162622. 84. Kufrin,D.,Eslin,D.E.,Bdeir,K.,Murciano,J.C.,Kuo,A.,Kowalska,M.A.,Degen, J.L., Sachais, B.S., Cines, D.B., and Poncz, M. 2003. Antithrombotic thrombocytes: ectopic expression of urokinasetype plasminogen activator in platelets. Blood 102:926933. 85. Ding, B.S., Hong, N., Murciano, J.C., Ganguly, K., Gottstein, C., Christofidou Solomidou, M., Albelda, S.M., Fisher, A.B., Cines, D.B., and Muzykantov, V.R. 2008. Prophylactic thrombolysis by thrombinactivated latent prourokinase targetedtoPECAM1inthepulmonaryvasculature. Blood 111:19992006. 86. Tavassoli, M. 1980. Megakaryocyteplatelet axis and the process of platelet formationandrelease. Blood 55:537545. 87. Sullivan, M.T., Cotten,R., Read, E.J., and Wallace, E.L. 2007. Bloodcollection andtransfusionintheUnitedStatesin2001. Transfusion 47:385394. 88. Choi, E.S., Nichol, J.L., Hokom, M.M., Hornkohl, A.C., and Hunt, P. 1995. Platelets generated in vitro from proplateletdisplaying human megakaryocytes arefunctional. Blood 85:402413. 89. Thornton,M.A.,Zhang,C.,Kowalska,M.A.,andPoncz,M.2002.Identificationof distal regulatory regions in the human alpha IIb gene locus necessary for consistent,highlevelmegakaryocyteexpression. Blood 100:35883596. 90. Wagner, C.L., Mascelli, M.A., Neblock, D.S., Weisman, H.F., Coller, B.S., and Jordan,R.E.1996.AnalysisofGPIIb/IIIareceptornumberbyquantificationof 7E3bindingtohumanplatelets. Blood 88:907914. 91. Lecine, P., Blank, V., and Shivdasani, R. 1998. Characterization of the hematopoietictranscriptionfactorNFE2inprimarymurinemegakaryocytes. J Biol Chem 273:75727578. 92. Levine, R.F., and Fedorko, M.E. 1976. Isolation of intact megakaryocytes from guineapigfemoralmarrow.Successfulharvestmadepossiblewithinhibitionsof plateletaggregation;enrichmentachievedwithatwostepseparationtechnique. J Cell Biol 69:159172. 93. Shivdasani,R.A.,andSchulze,H.2005.Culture,expansion,anddifferentiationof murinemegakaryocytes. Curr Protoc Immunol Chapter22:Unit22F26. 94. Brecher, G., and Cronkite, E.P. 1950. Morphology and enumeration of human bloodplatelets. J Appl Physiol 3:365377. 89

95. Kanaji,T.,Russell,S.,Cunningham,J.,Izuhara,K.,Fox,J.E.,andWare,J.2004. Megakaryocyte proliferation and ploidy regulated by the cytoplasmic tail of glycoproteinIbalpha. Blood 104:31613168. 96. Zhang,C.,Thornton,M.A.,Kowalska,M.A.,Sachis,B.S.,Feldman,M.,Poncz,M., McKenzie,S.E.,andReilly,M.P.2001.Localizationofdistalregulatorydomains inthemegakaryocytespecificplateletbasicprotein/plateletfactor4genelocus. Blood 98:610617. 97. Neyman, M., Gewirtz, J., and Poncz, M. 2008. Analysis of the spatial and temporal characteristics of plateletdelivered factor VIIIbased clots. Blood 112:11011108. 98. Blue,R.,Kowalska,M.A.,Hirsch,J.,Murcia,M.,Janczak,C.A.,Harrington,A., Jirouskova,M.,Li,J.,Fuentes,R.,Thornton,M.A., et al. 2009. Structural and therapeutic insights from the species specificity and in vivo antithrombotic activityofanovelalphaIIbspecificalphaIIbbeta3antagonist. Blood 114:195201. 99. JilmaStohlawetz, P., Homoncik, M., Jilma, B., Folman, C.C., von dem Borne, A.E.,Bernaschek,G.,Deutinger,J.,Ulm,B.,Eppel,W.,andPanzer,S.2001.High levels of reticulated platelets and thrombopoietin characterize fetal thrombopoiesis. Br J Haematol 112:466468. 100. HsuLin,S.,Berman,C.L.,Furie,B.C.,August,D.,andFurie,B.1984.Aplatelet membrane protein expressed during platelet activation and secretion. Studies using a monoclonal antibody specific for thrombinactivated platelets. J Biol Chem 259:91219126. 101. Peng, J., Friese, P., Heilmann, E., George, J.N., Burstein, S.A., and Dale, G.L. 1994.AgedplateletshaveanimpairedresponsetothrombinasquantitatedbyP selectinexpression. Blood 83:161166. 102. Rinder,H.M.,Murphy,M.,Mitchell,J.G.,Stocks,J.,Ault,K.A.,andHillman,R.S. 1991.Progressiveplateletactivationwithstorage:evidenceforshortenedsurvival ofactivatedplateletsaftertransfusion. Transfusion 31:409414. 103. Bergmeier, W., Burger, P.C., Piffath, C.L., Hoffmeister, K.M., Hartwig, J.H., Nieswandt,B.,andWagner,D.D.2003.Metalloproteinaseinhibitorsimprovethe recovery and hemostatic function of in vitroaged or injured mouse platelets. Blood 102:42294235. 104. Bergmeier,W.,Piffath,C.L.,Cheng,G.,Dole,V.S.,Zhang,Y.,vonAndrian,U.H., and Wagner, D.D. 2004. Tumor necrosis factoralphaconverting enzyme (ADAM17)mediatesGPIbalphasheddingfromplateletsinvitroandinvivo. Circ Res 95:677683. 105. Kahr,W.H.,Zheng,S.,Sheth,P.M.,Pai,M.,Cowie,A.,Bouchard,M.,Podor,T.J., Rivard, G.E., and Hayward, C.P. 2001. Platelets frompatients with theQuebec platelet disorder contain and secrete abnormal amounts of urokinasetype plasminogenactivator. Blood 98:257265. 106. Schulze, H., and Shivdasani, R.A. 2005. Mechanisms of thrombopoiesis. J Thromb Haemost 3:17171724. 107. Nichols,K.E.,Crispino,J.D.,Poncz,M.,White,J.G.,Orkin,S.H.,Maris,J.M.,and Weiss, M.J. 2000. Familial dyserythropoietic anaemia and thrombocytopenia duetoaninheritedmutationinGATA1. Nat Genet 24:266270. 108. Cantor, A.B. 2005. GATA transcription factors in hematologic disease. Int J Hematol 81:378384. 90

109. Steensma,D.P.,Dewald,G.W.,Lasho,T.L.,Powell,H.L.,McClure,R.F.,Levine, R.L., Gilliland, D.G., and Tefferi, A. 2005. The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both "atypical" myeloproliferative disordersandmyelodysplasticsyndromes. Blood 106:12071209. 110. Houwerzijl,E.J.,Blom,N.R.,vanderWant,J.J.,Esselink,M.T.,Koornstra,J.J., Smit, J.W., Louwes, H., Vellenga, E., and de Wolf, J.T. 2004. Ultrastructural study shows morphologic features of apoptosis and paraapoptosis in megakaryocytesfrompatientswithidiopathicthrombocytopenicpurpura. Blood 103:500506. 111. DunoisLarde, C., Capron, C., Fichelson, S., Bauer, T., CramerBorde, E., and Baruch, D. 2009. Exposure of human megakaryocytes to high shear rates acceleratesplateletproduction. Blood 114:18751883. 112. Schmitt, A., Guichard, J., Masse, J.M., Debili, N., and Cramer, E.M. 2001. Of miceandmen:comparisonoftheultrastructureofmegakaryocytesandplatelets. Exp Hematol 29:12951302. 113. Weibel, E.R., and Gomez, D.M. 1962. Architecture of the human lung. Use of quantitativemethodsestablishesfundamentalrelationsbetweensizeandnumber oflungstructures. Science 137:577585. 114. Davis,R.E.,Stenberg, P.E.,Levin, J., and Beckstead, J.H.1997.Localizationof megakaryocytes in normal mice and following administration of platelet antiserum, 5fluorouracil, or radiostrontium: evidence for the site of platelet production. Exp Hematol 25:638648. 115. Lill,M.C.,Perloff,J.K.,andChild,J.S.2006.Pathogenesisofthrombocytopenia incyanoticcongenitalheartdisease. Am J Cardiol 98:254258. 116. Dickinson, C.J. 1993. The aetiology of clubbing and hypertrophic osteoarthropathy. Eur J Clin Invest 23:330338. 117. Forestier,F.,Daffos,F.,Galacteros,F.,Bardakjian,J.,Rainaut,M.,andBeuzard, Y.1986.Hematologicalvaluesof163normalfetusesbetween18and30weeksof gestation. Pediatr Res 20:342346. 118. Baker, G.R.,Sullam, P.M., and Levin, J. 1997.A simple, fluorescentmethod to internallylabelplateletssuitableforphysiologicalmeasurements. Am J Hematol 56:1725. 119. Knust, J., Ochs, M., Gundersen, H.J., and Nyengaard, J.R. 2009. Stereological estimates of alveolar number and size and capillary length and surface area in micelungs. Anat Rec (Hoboken) 292:113122. 120. Nishikii,H.,Eto,K.,Tamura,N.,Hattori,K.,Heissig,B.,Kanaji,T.,Sawaguchi, A., Goto, S., Ware, J., and Nakauchi, H. 2008. Metalloproteinase regulation improvesinvitrogenerationofefficaciousplateletsfrommouseembryonicstem cells. J Exp Med 205:19171927. 121. Stachura, D.L., Chou, S.T., and Weiss, M.J. 2006. Early block to erythromegakaryocytic development conferred by loss of transcription factor GATA1. Blood 107:8797. 122. Yarovoi,H.V.,Kufrin,D.,Eslin,D.E.,Thornton,M.A.,Haberichter,S.L.,Shi,Q., Zhu,H.,Camire,R.,Fakharzadeh,S.S.,Kowalska,M.A.,etal.2003.FactorVIII ectopically expressed in platelets: efficacy in hemophilia A treatment. Blood 102:40064013. 123. Bradberry,J.C. 2004. Peripheral arterial disease: pathophysiology, riskfactors, androleofantithrombotictherapy. J Am Pharm Assoc (2003) 44:S3744;quiz S4435. 91

124. Bougie,D.W.,Birenbaum,J.,Rasmussen,M.,Poncz,M.,andAster,R.H.2009. Quininedependent, plateletreactive monoclonals mimic antibodies found in patientswithquinineinducedimmunethrombocytopenia. Blood 113:11051111. 125. Ding,B.S.,Gottstein,C.,Grunow,A.,Kuo,A.,Ganguly,K.,Albelda,S.M.,Cines, D.B., and Muzykantov, V.R. 2005. Endothelial targeting of a recombinant constructfusingaPECAM1singlechainvariableantibodyfragment(scFv)with prourokinasefacilitatesprophylacticthrombolysisinthepulmonaryvasculature. Blood 106:41914198. 126. Zaitsev, S., Spitzer, D., Murciano, J.C., Ding, B.S., Tliba, S., Kowalska, M.A., MarcosContreras, O.A., Kuo, A., Stepanova, V., Atkinson, J.P., et al. 2010. Sustained thromboprophylaxis mediated by an RBCtargeted prourokinase zymogenactivatedatthesiteofclotformation. Blood 115:52415248. 127. Dubel, S., Breitling, F., Fuchs, P., Zewe, M., Gotter, S., Welschof, M., Moldenhauer, G., and Little, M. 1994. Isolation of IgG antibody FvDNA from variousmouseandrathybridomacelllinesusingthepolymerasechainreaction withasimplesetofprimers. J Immunol Methods 175:8995. 128. Holliger,P.,Prospero,T.,andWinter,G.1993."Diabodies":smallbivalentand bispecificantibodyfragments. Proc Natl Acad Sci U S A 90:64446448. 129. Wang,Y.X.,MartinMcNulty,B.,Freay,A.D.,Sukovich,D.A.,HalksMiller,M., Li, W.W., Vergona, R., Sullivan, M.E., Morser, J., Dole, W.P., et al. 2001. Angiotensin II increases urokinasetype plasminogen activator expression and inducesaneurysmintheabdominalaortaofapolipoproteinEdeficientmice. Am J Pathol 159:14551464. 130. Simak, J., Holada, K., Janota, J., and Stranak, Z. 1999. Surface expression of majormembraneglycoproteinsonrestingandTRAPactivatedneonatalplatelets. Pediatr Res 46:445449. 131. Verheul,H.M.,Jorna,A.S.,Hoekman,K.,Broxterman,H.J.,Gebbink,M.F.,and Pinedo, H.M. 2000. Vascular endothelial growth factorstimulated endothelial cellspromoteadhesionandactivationofplatelets. Blood 96:42164221. 132. Majka, M., JanowskaWieczorek, A., Ratajczak, J., Kowalska, M.A., Vilaire, G., Pan,Z.K.,Honczarenko,M.,Marquez,L.A.,Poncz,M.,andRatajczak,M.Z.2000. Stromalderivedfactor1andthrombopoietinregulatedistinctaspectsofhuman megakaryopoiesis. Blood 96:41424151. 133. Wall,J.E.,BuijsWilts,M.,Arnold,J.T.,Wang,W.,White,M.M.,Jennings,L.K., and Jackson, C.W. 1995. A flow cytometric assay using mepacrine for study of uptake and release of platelet dense granule contents. Br J Haematol 89:380 385. 134. Lambert, M.P., Rauova, L., Bailey, M., SolaVisner, M.C., Kowalska, M.A., and Poncz, M. 2007. Platelet factor 4 is a negative autocrine in vivo regulator of megakaryopoiesis:clinicalandtherapeuticimplications. Blood 110:11531160. 135. Zaitsev, S., Spitzer, D., Murciano, J.C., Ding, B.S., Tliba, S., Kowalska, M.A., Bdeir, K., Kuo, A., Stepanova, V., Atkinson, J.P., et al. 2010. Targeting of a mutant plasminogen activator to circulating red blood cells for prophylactic fibrinolysis. J Pharmacol Exp Ther 332:10221031. 136. Ganguly, K., Krasik, T.,Medinilla, S., Bdeir, K.,Cines, D.B., Muzykantov, V.R., and Murciano, J.C. 2005. Blood clearance and activity of erythrocytecoupled fibrinolytics. J Pharmacol Exp Ther 312:11061113. 137. Carmeliet, P., Stassen, J.M., Schoonjans, L., Ream, B., van den Oord, J.J., De Mol,M.,Mulligan,R.C.,andCollen,D.1993.Plasminogenactivatorinhibitor1 genedeficient mice. II.Effectsonhemostasis,thrombosis, and thrombolysis. J Clin Invest 92:27562760. 92

138. Suzuki,Y.,Nagai,N.,andCollen,D.2004.Comparativeeffectsofmicroplasmin andtissuetypeplasminogenactivator(tPA)oncerebralhemorrhageinamiddle cerebralarteryocclusionmodelinmice. J Thromb Haemost 2:16171621. 139. Murciano, J.C., Medinilla, S., Eslin, D., Atochina, E., Cines, D.B., and Muzykantov,V.R.2003.Prophylacticfibrinolysisthroughselectivedissolutionof nascentclotsbytPAcarryingerythrocytes. Nat Biotechnol 21:891896. 140. Runge,M.S.,Harker,L.A.,Bode,C.,Ruef,J.,Kelly,A.B.,Marzec,U.M.,Allen,E., Caban, R., Shaw, S.Y., Haber, E., et al. 1996. Enhanced thrombolytic and antithrombotic potency of a fibrintargeted plasminogen activator in baboons. Circulation 94:14121422. 141. Benedict,C.R.,Refino,C.J.,Keyt,B.A.,Pakala,R.,Paoni,N.F.,Thomas,G.R.,and Bennett, W.F. 1995.New variant ofhuman tissue plasminogen activator (TPA) with enhanced efficacy and lower incidence of bleeding compared with recombinanthumanTPA. Circulation 92:30323040. 142. Chapman,D.F.,Lyden,P.,Lapchak,P.A.,Nunez,S.,Thibodeaux,H.,andZivin,J. 2001.ComparisonofTNKwithwildtypetissueplasminogenactivatorinarabbit embolicstrokemodel. Stroke 32:748752. 143. Marder,V.J.,andStewart,D.2002.Towardssaferthrombolytictherapy. Semin Hematol 39:206216. 144. Thomas,G.R.,Thibodeaux,H.,Errett,C.J.,Badillo,J.M.,Keyt,B.A.,Refino,C.J., Zivin, J.A., and Bennett, W.F. 1994. A longhalflife and fibrinspecific form of tissueplasminogenactivatorinrabbitmodelsofembolicstrokeandperipheral bleeding. Stroke 25:20722078;discussion20782079. 145. Zhang,R.L.,Zhang,L.,Jiang,Q.,Zhang,Z.G.,Goussev,A.,andChopp,M.2000. Postischemic intracarotid treatment with TNKtPA reduces infarct volume and improvesneurologicaldeficitsinembolicstrokeintheunanesthetizedrat. Brain Res 878:6471. 146. Zaitsev, S., Danielyan, K., Murciano, J.C.,Ganguly, K., Krasik, T., Taylor, R.P., Pincus, S., Jones, S., Cines, D.B., and Muzykantov, V.R. 2006. Human complementreceptortype1directedloadingoftissueplasminogenactivatoron circulatingerythrocytesforprophylacticfibrinolysis. Blood 108:18951902. 147. Danielyan, K., Ganguly, K., Ding, B.S., Atochin, D., Zaitsev, S., Murciano, J.C., Huang, P.L., Kasner, S.E., Cines, D.B., and Muzykantov, V.R. 2008. Cerebrovascularthromboprophylaxisinmicebyerythrocytecoupledtissuetype plasminogenactivator. Circulation 118:14421449. 148. Topol, E.J., Morris, D.C., Smalling, R.W., Schumacher, R.R., Taylor, C.R., Nishikawa, A., Liberman, H.A., Collen, D., Tufte, M.E., Grossbard, E.B., et al. 1987. A multicenter, randomized, placebocontrolled trial of a new form of intravenous recombinant tissuetype plasminogen activator (activase) in acute myocardialinfarction. J Am Coll Cardiol 9:12051213. 149. Kwiatkowski,T.G.,Libman,R.B.,Frankel,M.,Tilley,B.C.,Morgenstern,L.B.,Lu, M., Broderick, J.P., Lewandowski, C.A., Marler, J.R., Levine, S.R., et al. 1999. Effects of tissue plasminogen activator for acute ischemic stroke at one year. National Institute of Neurological Disorders and Stroke Recombinant Tissue PlasminogenActivatorStrokeStudyGroup. N Engl J Med 340:17811787. 150. Hayward,C.P.,Cramer,E.M.,Kane,W.H.,Zheng,S.,Bouchard,M.,Masse,J.M., and Rivard, G.E. 1997. Studies of a second family with the Quebec platelet disorder:evidencethatthedegradationofthealphagranulemembraneandits soluble contents are not secondary to a defect in targeting proteins to alpha granules. Blood 89:12431253. 93

151. Hayward,C.P.,Rivard,G.E.,Kane,W.H.,Drouin,J.,Zheng,S.,Moore,J.C.,and Kelton, J.G. 1996. An autosomal dominant, qualitative platelet disorder associated with multimerin deficiency, abnormalities in platelet factor V, thrombospondin, von Willebrand factor, and fibrinogen and an epinephrine aggregationdefect. Blood 87:49674978. 152. Hayward, C.P., Welch, B., Bouchard, M., Zheng, S., and Rivard, G.E. 1997. Fibrinogen degradation products in patients with the Quebec platelet disorder. Br J Haematol 97:497503. 153. McKay,H.,Derome,F.,Haq,M.A.,Whittaker,S.,Arnold,E.,Adam,F.,Heddle, N.M., Rivard, G.E., and Hayward, C.P. 2004. Bleeding risks associated with inheritanceoftheQuebecplateletdisorder. Blood 104:159165. 154. Muller,D.W.,Topol,E.J.,Ellis,S.G.,Sigmon,K.N.,Lee,K.,andCaliff,R.M.1991. Multivesselcoronaryarterydisease:akeypredictorofshorttermprognosisafter reperfusion therapy for acute myocardial infarction. Thrombolysis and AngioplastyinMyocardialInfarction(TAMI)StudyGroup. Am Heart J 121:1042 1049. 155. Ding, B.S., Hong, N., ChristofidouSolomidou, M., Gottstein, C., Albelda, S.M., Cines, D.B., Fisher, A.B., and Muzykantov, V.R. 2009. Anchoring fusion thrombomodulin to theendothelial lumenprotects against injuryinduced lung thrombosisandinflammation. Am J Respir Crit Care Med 180:247256. 156. Valerius, T., Stockmeyer, B., van Spriel, A.B., Graziano, R.F., van den Herik Oudijk, I.E.,Repp, R.,Deo, Y.M.,Lund,J.,Kalden, J.R., Gramatzki, M., et al. 1997. FcalphaRI (CD89) as a novel trigger molecule for bispecific antibody therapy. Blood 90:44854492.

94