1
Materials and Methods • Mice: 6-8 week old female C57BL/6 • Tumor cell line: Syngeneic B16F0 melanoma injected sc into both flanks • Adenovirus vector: Replication-incompetent Ad5 • Vector for assessing cellular uptake: Ad5-CMV-GFP • Vector for inducing mIL-12: Ad5 containing gene construct for mIL-12 under control of RTS (Ad-RTS-mIL-12) • Administration of adenovectors: 10 10 viral particles (vp) injected intra-tumorally (i.t.) at 11D, when tumors were palpable • Activator ligand (AL): 1000 mg/kg chow; or 2-35 mg/kg body weight in Labrasol, by oral gavage for gene expression and cytokines in tumor or serum • Phase 1b clinical trial with DC-RTS-Il-12 i.t.+ AL (0.6-200mg orally daily for 14 days for 1-5 treatment cycles Mechanism of Action of Therapy of B16F0 Melanoma in Mice, with DC-RTS-mIL-12 i.t. + oral AL • Conditional interleukin-12 gene therapy promotes safe and effective antitumor immunity H Komita, X Zhao, AK Katakam, P Kumar, M Kawabe, H Okada, JM Braughler and WJ Storkus. Cancer Gene Therapy (2009), 1–9 DC-RTS-mIL-12 injected intratumorally (i.t.) into B16F0 + AL induced sytemic anti- tumor immunity and regression of B16 tumors DC-RTS-mIL-12 + AL had prolonged survival in tumor and draining lymph nodes Treatment efficacy correlated with systemic anti-B16 CD8+ T cells in ELISPOT assay Recent studies: Mice treated with DC-RTS-IL-12 + AL had increased numbers of tumor-specific T cells in both tumor-draining lymph nodes and spleen capable of secreting granzyme B (GzB) and IFNγ in response to both CD8+ and CD4+ tumor peptides measured by ELISPOT. Mechanism of Action of Therapy of Patients with Advanced Stage Melanoma, with DC-RTS-hIL-12 i.t. + oral AL • Measurement of sera for levels of IL-12 and IL-12 regulated cytokines, by Luminex assay, indicated significant increase in IL-12 and/or IFNγ only after AL at ≥60 mg/day, even though some patients showed clinical disease control at substantially lower doses of AL • ELISPOT assays of IFNγ secretion indicated increased responses of both CD8+ and CD4+ T cells after stimulation with melanoma-specific peptides • In tumor biopsies after treatment: Increased number of CD4+ and/or CD8+ T cells No increase in T regulatory cells Decrease in myeloid-derived suppressor cells in some patients • Details of this trial will be presented at the ASCO annual meeting in June 2011 RheoSwitch ® + AL Controls Timing and Level of Target Gene Expression • The RheoSwitch Therapeutic System (RTS) contains three basic components: an inducible promoter a ligand-inducible transcription factor and a co-activation partner a RheoSwitch activator ligand (AL) • In the absence of ligand, the switch protein complex provides an “off” signal • In the presence of ligand, the complex changes conformation and provides a dose=dependent “on” signal for target gene expression In vivo, the orally administered AL turns on gene expression within 24 hours, and upon withdrawal of the AL, gene expression returns to baseline levels within about 24 hours Hypothesized Therapeutic Mechanism of Action of RTS-IL-12 + AL (1) RTS- IL-12 construct is inserted intoadenoviral vector backbone,( Ad-RTS-IL-12). (2) Syngeneic mouse splenocytes, or autologous blood monocytes isolated and purified by leukapheresis and elutriation, are differentiated ex vivo into dendritic cells (DCs) (3) DCs are transduced ex vivo with Ad- RTS-IL-12. (4) ~5x10 7 transduced DCs are injected into one or more target tumor lesions. (5) Alternatively, Ad-RTS-IL-12I viral construct from #1 above can be directly injected into target tumors. (6) Oral AL is administered at dose(s) to regulate desired timing and level of expression of IL-12 and IL-12 associated genes in tumor. (7) IL-12 drives DCs in tumor to develop into DC1s, which preferentially present tumor antigens to TH1 cells (8) This induces specific anti-tumor CD4+ TH1 cells and CD8+ CTL, locally and then systemically Other Poster Presentation on this Therapeutic Approach During 2011 Annual ASGCT Meeting: Poster 883 (May 19, 2011): Murugesan et al, Rheoswitch-Mediated Regulation of IL-12 Protein Delivered Using an Adenoviral Vector Results in Anti-Tumor Effects Across a Spectrum of Tumor Types That poster was focused on the therapeutic efficacy of Ad-RTS-mIL-12 + AL, and is complementary to this poster, which focuses on the immunologic mechanism of action (MOA) of this therapeutic approach, and on the analogous MOA studies in B16-tumor- bearing mice and melanoma patients with DCs transduced with Ad-RTS-IL-12 + AL Intratumoral Injection of Ad-RTS-IL-12 + AL Treatment Modulates Lymphoid and Myelomonocytic Phenotypes within B16F0 Tumors at Day 7 Cell Type Tumor Treatment Ad-RTS-mIL-12 %+ Ad-RTS-mIL-12 + AL %+ CD3+, CD4+ 4.7 10.9 CD3+, CD8+ 10.5 26.5 CD3-, NK1.1+ 0 7.9 B200+ 9.9 4.2 CD19+ 6.6 2.2 CD14+ 3.1 18.1 F480+ 19.0 28.7 CD11c+ 21.7 19.9 Local and Systemic Anti-Tumor Immunity is Induced by Rheoswitch Regulated IL-12 Production After Intra-Tumoral Injection of Adenovirus Vector as Well as Vector-Transduced Dendritic Cells ZIOPHARM Oncology, Inc. 1 First Avenue, Parris Building #34, Navy Yard Plaza, Boston, MA 02129 Main 617-259-1970 Fax 617-241-2855 www.ziopharm.com Cellular Uptake of Ad-CMV-GFP Directly Injected into Subcutaneous B16F0 Tumors % GFP+ Cells Intra-tumoral injection with Ad- CMV-GFP (Time after injection) Tumor (CD44+) Cells Host infiltrating (CD45+) Cells None 0 0 12 h 3.5 2.5 36 h 14.5 2.7 7 days 0 0 mIL12 and mIFNγ RNA Expression in B16 Melanoma After AL + i.t. Injection with Ad-RTS-mIL-12 mIL12 and mIL12 Regulated Cytokine Levels in B16 Tumor After AL + i.t. Injection with Ad-RTS-mIL-12 mIL12 and mIL12 Regulated Cytokine Levels in Serum After Treatment of B16 Tumor-Bearing Mice with AL + i.t. Ad-RTS-mIL-12 Granzyme B ELISPOT Assays in Response to Melanoma-Specific CD8+ Restricted Peptides at 7 Days After Treatment of B16 Tumor-Bearing Mice with Ad-RTS-mIL-12 + AL IFNγ ELISPOT Assays in Response to Melanoma- Specific CD8+ or CD4+ (TRP-1) Restricted Peptides at 7 Days After Treatment of B16 Tumor-Bearing Mice with Ad-RTS-mIL-12 + AL IFNγ ELISPOT Assays in Response to Melanoma- Specific CD8+ or CD4+ (TRP-1) Restricted Peptides at 7 Days After Treatment of B16 Tumor-Bearing Mice with Ad-RTS-mIL-12 + AL • Treatment of B16F0-bearing mice with AL + i.t. injection with DC-RTS-IL-12 induces systemic anti-melanoma immunity, including CTL • Treatment of B16F0-bearing mice with direct i.t. injection with Ad-RTS-IL-12 vector also induces systemic anti-melanoma immunity, with similar if not identical mechanism of action: Macrophages and plasmacytoid dendritic/myeloid cells as well as T cells and mela- noma take up Ad vector within 12 hours after i.t. injection By 7 days after i.t. Ad-RTS-IL-12 + AL, there is a shift to increased percentages of CD4+ & CD8+ T cells, NK cells, and macrophages By 1 day after treatment with AL at intermediate dose as well as high dose, expression of gene for IL-12 is increased and is still increased at day 7, and IFNγ gene expression increases by day 2 and is high at day 7 IL-12 in the injected tumor and in serum is mainly observed after high dose dose of AL, whereas other IL-12 associated cytokines are observed with even low dose of AL • Similar mechanism of action as well as therapeutic efficacy for i.t. treatment with Ad-RTS-IL-12 + AL or DC-RTS-IL-12 indicates a good basis for clinical trial with Ad-RTS-IL-12 + AL, which is planned for initiation in June 2011 Detection by PCR of Ad-RTS-mIL-12 DNA in Murine Immune Tissues, at 7 Days after Intra-Tumoral Injection of Ad-RTS-mIL-12 Tissue Treatment Sham (PBS) Injection Intra-Tumorally Intra-Tumoral Ad-RTS-mIL-12 Tumor + Spleen Tumor-Draining Lymph Nodes + Non-Draining Lymph Nodes 1 Intrexon Corp, Germantown, MD; 2 University of Pittsburgh Cancer Institute, Pittsburgh, PA; 3 ZIOPHARM Oncology, Inc., Boston, MA. Ronald B. Herberman 1 , Meixia Bi 1 , Mario Moreno 1 , Lisa Butterfield², Mary Jo Buffo², Mark O. Thornton 3 , and Kimberly A. Shafer-Weaver 1 . Poster #899 Abstract Interleukin-12 (IL-12) has been shown to be a key cytokine that enhances the ability of dendritic cells (DCs) to elicit anti-tumor T cells including TH1 cells and cytotoxic T lymphocytes (CTLs) as well as activation of natural killer (NK) cells. Although this cytokine is a potent mediator of anti-tumor immunity, its use in immunotherapy has been impeded by substantial toxicity due to administration of the recombinant protein. Komita et al [Cancer Gene Therapy (2009)] reported that gene therapy with intra-tumoral injection of syngeneic DCs transduced by an adenovirus vector, Ad-RTS-mIL-12, with the mIL-12 gene under the control of the Rheoswitch Therapeutic System(RTS) ], led to therapeutic efficacy against B16 melanoma. The mechanism of action appeared to be induction of systemic anti-tumor immunity, with CD8+ T cells secreting increased levels of IFNγ in response to specific tumor cells, and the persistence and accumulation of DCs in treated tumors. Our recent studies have indicated that mice treated with DC-RTS-IL-12 + an oral activator ligand(AL) had increased numbers of tumor-specific T cells in both tumor-draining lymph nodes and spleen capable of secreting granzyme B (GzB) and IFNγ in response to both CD8+ and CD4+ tumor peptides measured by ELISPOT. Since studies from our team (Murugesan et al) have shown that direct intra-tumoral injection of Ad-RTS-mIL-12 + oral AL also led to therapeutic efficacy against B16 and other murine tumors types, studies have been performed to deter mine whether the mechanism of action is similar to that of DC-RTS-mIL-12 + AL. To evaluate cell types transduced by direct adenovirus injection, adenovirus expressing green fluorescent protein (GFP) under a strong constitutive promoter was injected into established B16F0 melanoma tumors, and uptake of the vector after intratumoral injection was mainly in DCs and tumor cells. At day 7 after intra-tumoral injection of Ad-RTS-mIL-12 + daily AL, increased percentages of T cell subsets, NK cells and DCs were observed in the tumor microenvironment, and cells isolated from the DLN and spleens of the treated animals showed increased number of GzB and IFNγ-secreting T cells in response to CD8+ and CD4+ melanoma peptides by ELISPOT analysis. Taken together, these data indicate that intra-tumoral injection of either Ad-RTS-mIL-12 or DCs transduced ex vivo by this vector, in combination with daily oral AL, induced changes in the tumor micro-environment that favor anti-tumor immune responses and systemic anti-tumor immunity. Initial results from a phase 1b clinical trial with DC-RTS-IL-12 + AL in patients with advanced stages of melanoma indicated induction of melanoma- specific CD4+ and CD8+ peripheral blood T cells, by ELISPOT, in 2 of 4 melanoma patients. The analogous immunological mechanism of action with Ad-RTS-IL-12 in the mouse tumor model provides a good rationale for now translating the logistically simpler approach of direct injection of the Ad vector, compared to generation of the patient-specific transduced DC product, into accessible tumor lesions in combination with AL for clinical therapy. RheoSwitch Protein 1 RheoSwitch Components RheoSwitch Protein 2 Inducible Promoter Target Gene Therapeutic Gene Expression is OFF RheoSwitch Components Co-Activator Protein Ligand Basal Transcription Proteins Therapeutic Gene Expression is ON Not actual activator (1) (2) (3) (4) (5) (6) (6) (7) (4)(5) (7) (6) 7) (7 (4 4)(5) Macrophages, Plasmacytoid Dendritic/yeloid, and T Cells are the Primary Leukocyte Subpopulation Transduced by Ad-CMV-GFP Directly Injected into Subcutaneous B16f0 Tumors Cell Type Time After Intra-Tumoral Injection 12 Hours 36 Hours % of Total Cell in the Tumor % GFP+ (within cell type) % of Total Cells % GFP+ T(CD3+) Cells 9.9 0.3 9.9 0.3 NK(NK1.1+) Cells 0 0 0.2 0.1 B or DC (B220+) Cells 2.0 0 2.4 0.1 Macrophages (CD14+) 0.1 0 18.1 2.1 Macrophages (F480+) 2.2 0.3 2.7 0.1 Dendritic (CD11c+) Cells 0.3 0 0.6 0.1 Plamacytoid Dendritic/My- eloid (CD317+) Cells 36.7 1.7 21.2 2.4 mlFNg 0 10 20 30 40 50 60 70 80 90 100 0 1 2 4 7 Tumor mIFNg expression (Normalized to HPRT1) Time point(days) PBS AdmIL12 AdmIL12+2mg ligand AdmIL12+10mg ligand AdmIL12+35mg

Local and Systemic Anti-Tumor Immunity is Induced by ... · tumor micro-environment that favor anti-tumor immune responses and systemic anti-tumor immunity. Initial results from a

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Local and Systemic Anti-Tumor Immunity is Induced by ... · tumor micro-environment that favor anti-tumor immune responses and systemic anti-tumor immunity. Initial results from a

Materials and Methods

•Mice:6-8weekoldfemaleC57BL/6

•Tumorcellline:SyngeneicB16F0melanomainjectedscintobothflanks

•Adenovirusvector:Replication-incompetentAd5

•Vectorforassessingcellularuptake:Ad5-CMV-GFP

•VectorforinducingmIL-12:Ad5containinggeneconstructformIL-12undercontrolofRTS(Ad-RTS-mIL-12)

•Administrationofadenovectors:1010viralparticles(vp)injectedintra-tumorally(i.t.)at11D,whentumorswerepalpable

•Activatorligand(AL):1000mg/kgchow;or2-35mg/kgbodyweightinLabrasol,byoralgavageforgeneexpressionandcytokinesintumororserum

•Phase1bclinicaltrialwithDC-RTS-Il-12i.t.+AL(0.6-200mgorallydailyfor14daysfor1-5treatmentcycles

Mechanism of Action of Therapy of B16F0 Melanoma in Mice, with DC-RTS-mIL-12 i.t. + oral AL

•Conditionalinterleukin-12genetherapypromotessafeandeffectiveantitumorimmunity

HKomita,XZhao,AKKatakam,PKumar,MKawabe,HOkada,JMBraughlerandWJStorkus.CancerGeneTherapy(2009),1–9

▲DC-RTS-mIL-12injectedintratumorally(i.t.)intoB16F0+ALinducedsytemicanti-tumorimmunityandregressionofB16tumors

▲DC-RTS-mIL-12+ALhadprolongedsurvivalintumoranddraininglymphnodes

▲Treatmentefficacycorrelatedwithsystemicanti-B16CD8+TcellsinELISPOTassay

•Recent studies:MicetreatedwithDC-RTS-IL-12+ALhadincreasednumbersoftumor-specificTcellsinbothtumor-draininglymphnodesandspleencapableofsecretinggranzymeB(GzB)andIFNγinresponsetobothCD8+andCD4+tumorpeptidesmeasuredbyELISPOT.

Mechanism of Action of Therapy of Patients with Advanced Stage Melanoma, with DC-RTS-hIL-12 i.t. + oral AL

•MeasurementofseraforlevelsofIL-12andIL-12regulatedcytokines,byLuminexassay,indicatedsignificantincreaseinIL-12and/orIFNγonlyafterALat≥60mg/day,eventhoughsomepatientsshowedclinicaldiseasecontrolatsubstantiallylowerdosesofAL

•ELISPOTassaysofIFNγsecretionindicatedincreasedresponsesofbothCD8+andCD4+Tcellsafterstimulationwithmelanoma-specificpeptides

•Intumorbiopsiesaftertreatment:

▲IncreasednumberofCD4+and/orCD8+Tcells

▲NoincreaseinTregulatorycells

▲Decreaseinmyeloid-derivedsuppressorcellsinsomepatients

•DetailsofthistrialwillbepresentedattheASCOannualmeetinginJune2011

RheoSwitch® + AL Controls Timing and Level of Target Gene Expression

•TheRheoSwitchTherapeuticSystem(RTS)containsthreebasiccomponents:

▲aninduciblepromoter

▲aligand-inducibletranscriptionfactorandaco-activationpartner

▲aRheoSwitchactivatorligand(AL)

•Intheabsenceofligand,theswitchproteincomplexprovidesan“off”signal

•Inthepresenceofligand,thecomplexchangesconformationandprovidesadose=dependent“on”signalfortargetgeneexpression

•In vivo,theorallyadministeredALturnsongeneexpressionwithin24hours,anduponwithdrawaloftheAL,geneexpressionreturnstobaselinelevelswithinabout24hours

Hypothesized Therapeutic Mechanism of Action of RTS-IL-12 + AL

(1)RTS-IL-12constructisinsertedintoadenoviralvectorbackbone,(Ad-RTS-IL-12).

(2)Syngeneicmousesplenocytes,orautologousbloodmonocytesisolatedandpurifiedbyleukapheresisandelutriation,aredifferentiatedexvivointodendriticcells(DCs)

(3)DCsaretransducedexvivowithAd-RTS-IL-12.

(4)~5x107transducedDCsareinjectedintooneormoretargettumorlesions.

(5)Alternatively,Ad-RTS-IL-12Iviralconstructfrom#1abovecanbedirectlyinjectedintotargettumors.

(6)OralALisadministeredatdose(s)toregulatedesiredtimingandlevelofexpressionofIL-12andIL-12associatedgenesintumor.

(7) IL-12drivesDCsintumortodevelopintoDC1s,whichpreferentiallypresenttumorantigenstoTH1cells

(8)Thisinducesspecificanti-tumorCD4+TH1cellsandCD8+CTL,locallyandthensystemically

Other Poster Presentation on this Therapeutic Approach During 2011 Annual ASGCT Meeting:

Poster 883 (May 19, 2011): Murugesan et al, Rheoswitch-Mediated Regulation of IL-12 Protein Delivered Using an Adenoviral Vector Results in Anti-Tumor Effects Across a Spectrum of Tumor Types

ThatposterwasfocusedonthetherapeuticefficacyofAd-RTS-mIL-12+AL,andis

complementarytothisposter,whichfocusesontheimmunologicmechanismofaction

(MOA)ofthistherapeuticapproach,andontheanalogousMOAstudiesinB16-tumor-

bearingmiceandmelanomapatientswithDCstransducedwithAd-RTS-IL-12+AL

Intratumoral Injection of Ad-RTS-IL-12 + AL Treatment Modulates Lymphoid and Myelomonocytic Phenotypes within B16F0 Tumors at Day 7

Cell TypeTumor Treatment

Ad-RTS-mIL-12 %+

Ad-RTS-mIL-12 + AL%+

CD3+, CD4+ 4.7 10.9

CD3+, CD8+ 10.5 26.5

CD3-, NK1.1+ 0 7.9

B200+ 9.9 4.2

CD19+ 6.6 2.2

CD14+ 3.1 18.1

F480+ 19.0 28.7

CD11c+ 21.7 19.9

Local and Systemic Anti-Tumor Immunity is Induced by Rheoswitch Regulated IL-12 Production After Intra-Tumoral Injection of Adenovirus Vector as Well as Vector-Transduced Dendritic Cells

ZIOPHARM Oncology, Inc. 1FirstAvenue,ParrisBuilding#34,NavyYardPlaza,Boston,MA02129Main617-259-1970Fax617-241-2855www.ziopharm.com

Cellular Uptake of Ad-CMV-GFP Directly Injected into Subcutaneous B16F0 Tumors

% GFP+ Cells

Intra-tumoral injection with Ad-CMV-GFP

(Time after injection)Tumor (CD44+) Cells Host infiltrating (CD45+) Cells

None 0 0

12 h 3.5 2.5

36 h 14.5 2.7

7 days 0 0

mIL12 and mIFNγ RNA Expression in B16 Melanoma After AL + i.t. Injection with Ad-RTS-mIL-12

mIL12 and mIL12 Regulated Cytokine Levels in B16 Tumor After AL + i.t. Injection with Ad-RTS-mIL-12

mIL12 and mIL12 Regulated Cytokine Levels in Serum After Treatment of B16 Tumor-Bearing Mice with AL + i.t. Ad-RTS-mIL-12

Granzyme B ELISPOT Assays in Response to Melanoma-Specific CD8+ Restricted Peptides at 7 Days After Treatment of B16 Tumor-Bearing Mice with Ad-RTS-mIL-12 + AL

IFNγ ELISPOT Assays in Response to Melanoma-Specific CD8+ or CD4+ (TRP-1) Restricted Peptides at 7 Days After Treatment of B16 Tumor-Bearing Mice with Ad-RTS-mIL-12 + AL

IFNγ ELISPOT Assays in Response to Melanoma-Specific CD8+ or CD4+ (TRP-1) Restricted Peptides at 7 Days After Treatment of B16 Tumor-Bearing Mice with Ad-RTS-mIL-12 + AL

•TreatmentofB16F0-bearingmicewithAL+i.t.injectionwithDC-RTS-IL-12inducessystemicanti-melanomaimmunity,includingCTL

•TreatmentofB16F0-bearingmicewithdirecti.t.injectionwithAd-RTS-IL-12vectoralsoinducessystemicanti-melanomaimmunity,withsimilarifnotidenticalmechanismofaction:

▲Macrophagesandplasmacytoiddendritic/myeloidcellsaswellasTcellsandmela-nomatakeupAdvectorwithin12hoursafteri.t.injection

▲By7daysafteri.t.Ad-RTS-IL-12+AL,thereisashifttoincreasedpercentagesofCD4+&CD8+Tcells,NKcells,andmacrophages

▲By1dayaftertreatmentwithALatintermediatedoseaswellashighdose,expressionofgeneforIL-12isincreasedandisstillincreasedatday7,andIFNγgeneexpressionincreasesbyday2andishighatday7

▲IL-12intheinjectedtumorandinserumismainlyobservedafterhighdosedoseofAL,whereasotherIL-12associatedcytokinesareobservedwithevenlowdoseofAL

•Similarmechanismofactionaswellastherapeuticefficacyfori.t.treatmentwithAd-RTS-IL-12+ALorDC-RTS-IL-12indicatesagoodbasisforclinicaltrialwithAd-RTS-IL-12+AL,whichisplannedforinitiationinJune2011

Detection by PCR of Ad-RTS-mIL-12 DNA in Murine Immune Tissues, at 7 Days after Intra-Tumoral Injection of Ad-RTS-mIL-12

TissueTreatment

Sham (PBS) Injection Intra-Tumorally

Intra-TumoralAd-RTS-mIL-12

Tumor – +

Spleen – –

Tumor-Draining Lymph Nodes – +

Non-Draining Lymph Nodes – –

1IntrexonCorp,Germantown,MD;2UniversityofPittsburghCancerInstitute,Pittsburgh,PA;3ZIOPHARMOncology,Inc.,Boston,MA.

Ronald B. Herberman1, Meixia Bi1, Mario Moreno1, Lisa Butterfield², Mary Jo Buffo², Mark O. Thornton3, and Kimberly A. Shafer-Weaver1.

Poster#899

Abstract

Interleukin-12(IL-12)hasbeenshowntobeakeycytokinethatenhancestheability

ofdendriticcells(DCs)toelicitanti-tumorTcellsincludingTH1cellsandcytotoxic

Tlymphocytes(CTLs)aswellasactivationofnaturalkiller(NK)cells.Althoughthis

cytokineisapotentmediatorofanti-tumorimmunity,itsuseinimmunotherapy

hasbeenimpededbysubstantialtoxicityduetoadministrationoftherecombinant

protein.Komitaetal[CancerGeneTherapy(2009)]reportedthatgenetherapywith

intra-tumoralinjectionofsyngeneicDCstransducedbyanadenovirusvector,

Ad-RTS-mIL-12,withthemIL-12geneunderthecontroloftheRheoswitch

TherapeuticSystem(RTS)™],ledtotherapeuticefficacyagainstB16melanoma.The

mechanismofactionappearedtobeinductionofsystemicanti-tumorimmunity,with

CD8+TcellssecretingincreasedlevelsofIFNγinresponsetospecifictumorcells,

andthepersistenceandaccumulationofDCsintreatedtumors.Ourrecentstudies

haveindicatedthatmicetreatedwithDC-RTS-IL-12+anoralactivatorligand(AL)

hadincreasednumbersoftumor-specificTcellsinbothtumor-draininglymphnodes

andspleencapableofsecretinggranzymeB(GzB)andIFNγinresponsetobothCD8+andCD4+tumorpeptidesmeasuredbyELISPOT.Sincestudiesfromourteam

(Murugesanetal)haveshownthatdirectintra-tumoralinjectionofAd-RTS-mIL-12+

oralALalsoledtotherapeuticefficacyagainstB16andothermurinetumorstypes,

studieshavebeenperformedtodeterminewhetherthemechanismofactionis

similartothatofDC-RTS-mIL-12+AL.Toevaluatecelltypestransducedbydirect

adenovirusinjection,adenovirusexpressinggreenfluorescentprotein(GFP)under

astrongconstitutivepromoterwasinjectedintoestablishedB16F0melanoma

tumors,anduptakeofthevectorafterintratumoralinjectionwasmainlyinDCs

andtumorcells.Atday7afterintra-tumoralinjectionofAd-RTS-mIL-12+dailyAL,

increasedpercentagesofTcellsubsets,NKcellsandDCswereobservedinthe

tumormicroenvironment,andcellsisolatedfromtheDLNandspleensofthetreated

animalsshowedincreasednumberofGzBandIFNγ-secretingTcellsinresponsetoCD8+andCD4+melanomapeptidesbyELISPOTanalysis.Takentogether,these

dataindicatethatintra-tumoralinjectionofeitherAd-RTS-mIL-12orDCstransduced

exvivobythisvector,incombinationwithdailyoralAL,inducedchangesinthe

tumormicro-environmentthatfavoranti-tumorimmuneresponsesandsystemic

anti-tumorimmunity.Initialresultsfromaphase1bclinicaltrialwithDC-RTS-IL-12+

ALinpatientswithadvancedstagesofmelanomaindicatedinductionofmelanoma-

specificCD4+andCD8+peripheralbloodTcells,byELISPOT,in2of4melanoma

patients.TheanalogousimmunologicalmechanismofactionwithAd-RTS-IL-12in

themousetumormodelprovidesagoodrationalefornowtranslatingthelogistically

simplerapproachofdirectinjectionoftheAdvector,comparedtogenerationofthe

patient-specifictransducedDCproduct,intoaccessibletumorlesionsincombination

withALforclinicaltherapy.

RheoSwitchProtein 1

RheoSwitch Components

RheoSwitchProtein 2

InduciblePromoter

TargetGene

RheoSwitch Components Co-ActivatorProtein

Ligand

BasalTranscriptionProteins

Therapeutic Gene Expression is OFF

Therapeutic Gene Expression is ON

RheoSwitchProtein 1

RheoSwitch Components

RheoSwitchProtein 2

InduciblePromoter

TargetGene

RheoSwitch Components Co-ActivatorProtein

Ligand

BasalTranscriptionProteins

Therapeutic Gene Expression is OFF

Therapeutic Gene Expression is ON

RheoSwitchProtein 1

RheoSwitch Components

RheoSwitchProtein 2

InduciblePromoter

TargetGene

RheoSwitch Components Co-ActivatorProtein

Ligand

BasalTranscriptionProteins

Therapeutic Gene Expression is OFF

Therapeutic Gene Expression is ON

Not actual activator (1)

(2)

(3)

(4)

(5)

(6)

(6)

(7)

(4)(5)

(7)

(6)

7)(7

(44)(5)

Macrophages, Plasmacytoid Dendritic/yeloid, and T Cells are the Primary Leukocyte Subpopulation Transduced by Ad-CMV-GFP Directly Injected into Subcutaneous B16f0 Tumors

Cell Type

Time After Intra-Tumoral Injection

12 Hours 36 Hours

% of Total Cell in the Tumor

% GFP+ (within cell type) % of Total Cells % GFP+

T(CD3+) Cells 9.9 0.3 9.9 0.3

NK(NK1.1+) Cells 0 0 0.2 0.1

B or DC (B220+) Cells 2.0 0 2.4 0.1

Macrophages (CD14+) 0.1 0 18.1 2.1

Macrophages (F480+) 2.2 0.3 2.7 0.1

Dendritic (CD11c+) Cells 0.3 0 0.6 0.1

Plamacytoid Dendritic/My-eloid (CD317+) Cells 36.7 1.7 21.2 2.4

mlFNgmlFNg

0 10 20 30 40 50 60 70 80 90

100

0 1 2 4 7

Tum

or m

IFN

g ex

pre

ssio

n(N

orm

aliz

ed t

o H

PR

T1)

Time point(days)

PBS

AdmIL12

AdmIL12+2mg ligand

AdmIL12+10mg

ligand

AdmIL12+35mg