cancer immunotherapy with t cells carrying bispeci c receptors that mimic antibodies · research...

12
Research Article Cancer Immunotherapy with T Cells Carrying Bispecic Receptors That Mimic Antibodies Sarah Ahn 1,2 , Jingjing Li 3,4 , Chuang Sun 1,2 , Keliang Gao 3,4 , Koichi Hirabayashi 2 , Hongxia Li 2,5 , Barbara Savoldo 1,2,6 , Rihe Liu 2,3,4 , and Gianpietro Dotti 1,2 Abstract Tumors are inherently heterogeneous in antigen expres- sion, and escape from immune surveillance due to antigen loss remains one of the limitations of targeted immuno- therapy. Despite the clinical use of adoptive therapy with chimeric antigen receptor (CAR)redirected T cells in lym- phoblastic leukemia, treatment failure due to epitope loss occurs. Targeting multiple tumor-associated antigens (TAAs) may thus improve the outcome of CAR-T cell ther- apies. CARs developed to simultaneously target multiple targets are limited by the large size of each single-chain variable fragment and compromised protein folding when several single chains are linearly assembled. Here, we describe single-domain antibody mimics that function with- in CAR parameters but form a very compact structure. We show that antibody mimics targeting EGFR and HER2 of the ErbB receptor tyrosine kinase family can be assembled into receptor molecules, which we call antibody mimic receptors (amR). These amR can redirect T cells to recognize two different epitopes of the same antigen or two different TAAs in vitro and in vivo. Introduction Chimeric antigen receptors (CARs) are synthetic receptors. Their specicity is determined by the single-chain variable fragment (scFv) obtained from a monoclonal antibody, with their activation controlled by the z-chain signaling domain from the T-cell receptor complex and costimulatory endodomains (1, 2). Gene transfer of CARs into T cells redirects T-cell antigen specicity through the scFv. T-cell activation and proliferation is amplied through costi- mulatory signals (3). The infusion of CAR-T cells in patients with lymphoid malignancies has led to durable complete remission in more than 40% of patients (4, 5). However, up to 20% of patients with acute lymphoblastic leukemia receiving CD19-specic CAR-T cells relapse due to the emergence of leukemic clones that have lost the targeted epitope (4, 6). Furthermore, the heterogeneity of tumor-associated antigen (TAA) expression in solid tumors leads to CAR-T treatment failure when a single TAA is targeted (7). The generation of multiredirected CAR-T cells, namely, by recognition of two nonoverlapping epitopes of a TAA or two different TAAs, may be necessary to effectively eradicate tumor cells. Several approaches have been proposed to achieve this goal including pooling CAR-T products targeting different TAAs, generating vectors encoding two different CARs that can be expressed simultaneously by each T cell, and engineering cas- settes in which two scFvs are assembled into a single CAR moiety (2, 8, 9). Although these approaches are feasible, a challenge in using an scFv-based antigen receptor is the need for multiple V H and V L domains to appropriately pair and stabilize in complex structures. In addition to the complexity and costs of manufacturing multiple T-cell products, reduced expression of simultaneously expressed CARs, and compro- mised protein folding when several scFvs are linearly assembled in one single CAR, remain challenges to current methods of generating multiredirected CAR-T cells. Achieving multiple tumor-targeting features using scFv-based CAR-T cells is difcult. We hypothesized that extracellular antigen receptors with simple structure, high stability, and small size could address the challenge. Various small protein domains that are not structurally equivalent to the immunoglobulin domains have been developed using various display technologies (1014). Tumor- homing ligands based on a small protein domain possess advan- tages over those on a multidomain, complex structure in terms of engineering higher modularity. Ideally, the scaffold should be a monomeric small protein domain with high solubility and stability that is not inclined to aggregation, that tolerates sequence varia- tions, and that is amenable to directed molecular evolution to create antigen-binding ligands with multiple functions (13). Among numerous scaffolds that have been examined, three single-domain antibody mimics are of interest for developing CARs with multiple tumor-targeting features: (i) monobody, based on the type III domain of bronectin (FN3); (ii) afbody, based on a three-helix bundle Z domain; and (iii) DARPin, based on the designed ankyrin repeat protein (11, 12, 15, 16). These engineered proteins can have high specicity and afnity, despite their simple structures and relatively small size. The FN3 domain 1 Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. 2 Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. 3 Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. 4 Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. 5 Beijing Chest Hospital, Department of Medical Oncology, Capital Medical University, Beijing, China. 6 Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). Corresponding Authors: Gianpietro Dotti, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, 27514. Phone: 919-962-8414; Fax: 919-962- 8414; E-mail: [email protected]; Rihe Liu, Marsico Hall 3111; 125 Mason Farm Road Chapel Hill, NC 27599-7368; Phone: 919-843-3635; E-mail: [email protected] doi: 10.1158/2326-6066.CIR-18-0636 Ó2019 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org 773 on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

Upload: others

Post on 26-Jan-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Research Article

    Cancer Immunotherapy with T Cells CarryingBispecific Receptors That Mimic AntibodiesSarah Ahn1,2, Jingjing Li3,4, Chuang Sun1,2, Keliang Gao3,4, Koichi Hirabayashi2,Hongxia Li2,5, Barbara Savoldo1,2,6, Rihe Liu2,3,4, and Gianpietro Dotti1,2

    Abstract

    Tumors are inherently heterogeneous in antigen expres-sion, and escape from immune surveillance due to antigenloss remains one of the limitations of targeted immuno-therapy. Despite the clinical use of adoptive therapy withchimeric antigen receptor (CAR)–redirected T cells in lym-phoblastic leukemia, treatment failure due to epitopeloss occurs. Targeting multiple tumor-associated antigens(TAAs) may thus improve the outcome of CAR-T cell ther-apies. CARs developed to simultaneously target multipletargets are limited by the large size of each single-chain

    variable fragment and compromised protein folding whenseveral single chains are linearly assembled. Here, wedescribe single-domain antibody mimics that function with-in CAR parameters but form a very compact structure. Weshow that antibody mimics targeting EGFR and HER2 of theErbB receptor tyrosine kinase family can be assembled intoreceptor molecules, which we call antibody mimic receptors(amR). These amR can redirect T cells to recognize twodifferent epitopes of the same antigen or two different TAAsin vitro and in vivo.

    IntroductionChimeric antigen receptors (CARs) are synthetic receptors. Their

    specificity is determined by the single-chain variable fragment(scFv) obtained from amonoclonal antibody, with their activationcontrolled by the z-chain signaling domain from the T-cell receptorcomplex and costimulatory endodomains (1, 2). Gene transfer ofCARs into T cells redirects T-cell antigen specificity through thescFv. T-cell activation and proliferation is amplified through costi-mulatory signals (3). The infusion of CAR-T cells in patients withlymphoid malignancies has led to durable complete remission inmore than 40% of patients (4, 5). However, up to 20% of patientswith acute lymphoblastic leukemia receiving CD19-specific CAR-Tcells relapse due to the emergence of leukemic clones that have lostthe targeted epitope (4, 6). Furthermore, the heterogeneity oftumor-associated antigen (TAA) expression in solid tumors leadsto CAR-T treatment failure when a single TAA is targeted (7).

    The generation of multiredirected CAR-T cells, namely, byrecognition of two nonoverlapping epitopes of a TAA or two

    different TAAs, may be necessary to effectively eradicate tumorcells. Several approaches have been proposed to achieve thisgoal including pooling CAR-T products targeting different TAAs,generating vectors encoding two different CARs that can beexpressed simultaneously by each T cell, and engineering cas-settes in which two scFvs are assembled into a single CARmoiety (2, 8, 9). Although these approaches are feasible, achallenge in using an scFv-based antigen receptor is the needfor multiple VH and VL domains to appropriately pair andstabilize in complex structures. In addition to the complexityand costs of manufacturing multiple T-cell products, reducedexpression of simultaneously expressed CARs, and compro-mised protein folding when several scFvs are linearly assembledin one single CAR, remain challenges to current methods ofgenerating multiredirected CAR-T cells.

    Achieving multiple tumor-targeting features using scFv-basedCAR-T cells is difficult. We hypothesized that extracellular antigenreceptors with simple structure, high stability, and small size couldaddress the challenge. Various small protein domains that are notstructurally equivalent to the immunoglobulin domains have beendeveloped using various display technologies (10–14). Tumor-homing ligands based on a small protein domain possess advan-tages over those on a multidomain, complex structure in terms ofengineering higher modularity. Ideally, the scaffold should be amonomeric smallproteindomainwithhigh solubility and stabilitythat is not inclined to aggregation, that tolerates sequence varia-tions, and that is amenable to directed molecular evolution tocreate antigen-binding ligands with multiple functions (13).

    Among numerous scaffolds that have been examined, threesingle-domain antibody mimics are of interest for developingCARs with multiple tumor-targeting features: (i) monobody,based on the type III domain of fibronectin (FN3); (ii) affibody,based on a three-helix bundle Z domain; and (iii) DARPin, basedon the designed ankyrin repeat protein (11, 12, 15, 16). Theseengineered proteins can have high specificity and affinity, despitetheir simple structures and relatively small size. The FN3 domain

    1Department of Microbiology and Immunology, University of North Carolina atChapel Hill, Chapel Hill, North Carolina. 2Lineberger Comprehensive CancerCenter, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.3Eshelman School of Pharmacy, Division of Chemical Biology and MedicinalChemistry, University ofNorth Carolina at Chapel Hill, Chapel Hill, North Carolina.4Carolina Center for Genome Sciences, University of North Carolina at ChapelHill, Chapel Hill, North Carolina. 5Beijing Chest Hospital, Department of MedicalOncology, Capital Medical University, Beijing, China. 6Department of Pediatrics,University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.

    Note: Supplementary data for this article are available at Cancer ImmunologyResearch Online (http://cancerimmunolres.aacrjournals.org/).

    Corresponding Authors: Gianpietro Dotti, University of North Carolina at ChapelHill, 125 Mason Farm Road, Chapel Hill, 27514. Phone: 919-962-8414; Fax: 919-962-8414; E-mail: [email protected];Rihe Liu,MarsicoHall 3111; 125MasonFarmRoadChapel Hill, NC 27599-7368; Phone: 919-843-3635; E-mail: [email protected]

    doi: 10.1158/2326-6066.CIR-18-0636

    �2019 American Association for Cancer Research.

    CancerImmunologyResearch

    www.aacrjournals.org 773

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://crossmark.crossref.org/dialog/?doi=10.1158/2326-6066.CIR-18-0636&domain=pdf&date_stamp=2019-4-16http://cancerimmunolres.aacrjournals.org/

  • is a stable protein with a molecular weight (MW) around 10 kDa.Structurally, it has a b-sandwich scaffold similar to that of theimmunoglobulin VH domain, with putative ligand-binding sitescomposed of three solvent accessible surface loops that are struc-turally analogous to the CDR H1, H2, and H3 of the VHdomain (11). The advantage of the FN3 domain is in its lack ofdisulfide bonds and posttranslational modifications for biologicalfunctions. Affibodies (AFF) are based on the three-helix bundle Zdomain derived from Staphylococcal protein A (17, 18). As with theFN3 domain, AFF domains are resistant to proteolysis and heat-induced denaturation and lack disulfide bonds. Finally, DARPinscontain consecutive copies of small structural repeats, which stacktogether to form a contiguous interacting surface (14). DARPin-based targeting ligands that bind to various targets including CD4,EGFR, and HER2 have been generated (19).

    Taking into consideration the simplicity, stability, and smallersize of these targeting ligands, as well as their current applicationsin therapeutics and diagnostics (20), we explored the use of thesemolecules in generating antigen-specific receptors for T cells. Inparticular, we investigated if a combination of these single-domain antibodymimics allows the generation of a T-cell-surfaceantigen receptor that recognizes two different epitopes of thesame tumor antigen or two different antigens, aiming to developT cells with bispecific redirection targeting two epitopes of thesame antigen or two different antigens. As proof of principle, wehave adapted high-affinity antibody mimics specific for ErbB1(EGFR) and ErbB2 (HER2), to generate receptor molecules calledantibody mimic receptors (amR).

    Materials and MethodsConstruction of bispecific CAR vectors

    To construct bispecific CAR vectors, the codon-optimized (forexpression in human cells) coding regions for a monomeric orheterodimeric EGFR- or/and HER2-binding ligand were fusedthrough an optimized flexible linker. The final coding region wascloned into the SFG vector, resulting in a fusion protein that iscomposed of the signaling peptide from human IgG heavy-chain,EGFR- or HER2-binding domain(s), a FLAG tag, a 45-residuehinge region from the human CD8a extracellular domain, thetransmembrane domain of human CD8a, the CD28-costimula-tory endodomain, and the z chain of the TCR/CD3 complex (21).The CD8a hinge and transmembrane domains contain nativecysteine residues. Single-domain antibodymimics (AFF, DARPin,and FN3)were PCR amplified and cloned into the SFG vector. ThescFv derived from the cetuximab mAb was PCR amplified andcloned into the SFG vector. EGFR WT (Addgene plasmid#110110) and pBABE-puro-ErbB2 (Addgene plasmid #40978)were gifts from Matthew Meyerson Dana-Farber Cancer Institute.Full-length EGFR and HER2 were amplified by PCR and clonedinto the SFG retroviral vector. A truncated formofHER2 lacking anintracellulardomainwasamplifiedbyPCRandalso cloned into theSFG retroviral vector. All retroviral supernatants were prepared aspreviously described (22).

    Expression and purification of recombinant EGFR and HER2binding protein domains

    Coding sequences codon-optimized for expression in E. coliwith a C-terminal His tag were cloned into the pET28b vector. Toexpress the ligands, vectors were transformed into E. coli BL21(DE3) Rosetta cells, and positive clones were selected on lysogeny

    broth (LB) plates containing 50 mg/mL kanamycin and 34 mg/mLchloramphenicol. Single colonies were picked and grown over-night at 37�C. Overnight cell cultures were added to 1 L of LBmedia and grown at 37�C.When theOD600was between0.6 and0.8, 1mmol/L IPTGwas added to induce expression for 4 hours at37�C. To purify the binding ligands, the cell pellet was resus-pended in buffer A (25 mmol/L HEPES pH 7.4 and 300 mmol/LNaCl) supplemented with 1 mmol/L phenylmethylsulfonyl fluo-ride (PMSF) and sonicated on ice for 10minutes on a Sonifier 450sonicator (Branson). After cell lysis, the soluble fraction wasrecovered by centrifugation at 4�C. The resulting soluble fractionwas loaded onto an IMAC Ni-charged affinity column (Bio-Rad)preequilibrated with buffer A. The column was washed withbuffer A containing 20 mmol/L imidazole (buffer B) and then50 mmol/L imidazole (buffer C) and the proteins were elutedwith buffer D (buffer A and 200 mmol/L imidazole). Followingdialysis against 1 � PBS, the quality of the purified proteins wasverified by SDS-PAGE.

    Characterization of target-binding featuresBio-layer interferometry (BLI) analyses of the monomeric and

    heterodimeric EGFR andHER2-binding domainswere performedon a Octet QK system (ForteBio LLC.) at 30 �C. The ErbB bindingligands and corresponding receptors diluted to required concen-trations with an assay buffer (1 �PBS, 1% BSA, 0.05% Tween 20,pH 7.4) as well as the buffer are assigned to black 96 well plates(Greiner Bio-One). Streptavidin (SA) biosensors (Fort_eBio)wereused to immobilize biotinylated ErbB binding ligands and ErbBwith Fc fusions(AcroBiosystems) at three different concentrationsran against ligand binding SA sensors. Assays run in triplicatewereacquired and analyzed on the Fort�eBio Data Acquisition 6.4software. Savitzky–Golay filtering was applied to the averagedreference biosensors and then globally fitted at a 1:1 model.

    Cell linesTumor cell lines Panc-1, BxPC-3, HPAF-II, and AsPC-1 (pan-

    creatic cancer), MCF-7 (breast cancer), and BV173 (B cell lym-phoma) were purchased from ATCC. BxPC-3 and BV173 werecultured in RPMI-1640 (Gibco). AsPC-1 was cultured in RPMI-1640 supplemented with 1 mmol/L sodium pyruvate (Gibco).HPAF-II and MCF-7 were cultured in MEM (Gibco). Panc-1 wascultured in DMEM (Gibco). All media were supplemented with10% FBS (Sigma), 2 mmol/L GlutaMax (Gibco) and penicillin(100 units/mL) and streptomycin (100 mg/mL; Gibco). All cellswere maintained at 37�C with 5% CO2. All cell lines are regularlytested for Mycoplasma, and the identity of the cell lines wasvalidated by flow cytometry for relevant cell-surface markers andwere also monitored for morphological drift in culture. Cell lineswere maintained in culture no longer than 30 days and thenreplaced with cells from stored vials. The number of previouspassages of these cell lines was unknown. Panc-1 cells weretransduced with a retroviral vector encoding the eGFP-Firefly-Luciferase (eGFP-FFluc) gene (21). BV173 cells were transducedwith a retroviral construct encoding full-length human EGFRto generate BV173-EGFR cells or a truncated form of HER2 togenerate BV173-HER2 cells. Panc-1 cells were transduced with aretroviral vector encoding HER2 to make Panc-1-HER2.

    Generation of redirected T cellsT cells expressing CAR and amRs were generated in

    accordance with standard operating procedures currently used

    Ahn et al.

    Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research774

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • to manufacture CAR-T cells for clinical use at our institu-tion (24, 25). Peripheral blood mononuclear cells were isolatedfrom discharged buffy coats (Gulf Coast Regional Blood Center)using Lymphoprep medium (Accurate Chemical and ScientificCorporation) and activated on plates coated with 1 mg/mL CD3(Miltenyi Biotec) and CD28 (BD Biosciences) mAbs. ActivatedT cells were transduced with retroviral supernatants on retro-nectin-coated 24-well plates (Takara Bio Inc.) 2 to 3 days afteractivation. Transduced T cells were expanded in 50% Click'sMedium (Irvine Scientific) and 50% RPMI-1640 supplementedwith 10% HyClone FBS (GE Healthcare), 2 mmol/L GlutaMax(Gibco) and penicillin (100 units/mL) and streptomycin(100 mg/mL; Gibco) with 10 ng/mL IL7 and 5 ng/mL of IL15(PeproTech) for 10 to 14 days of culture before being used forfunctional assays (24–26).

    Flow cytometryWe used mAbs specific for human CD3 (APC-H7; SK7;

    560176), CD45 (BV510; HI30; 563204), CD4 (BV711; SK3;563028), CD8 (APC; SK1; 340584), CD19 (FITC; SJ25C1;340409), CD45RA (PE; HI100; 555489), CD45RO (BV786;UCHL1; 564290), CD69 (FITC; L78; 347823), and HER2 (PE;Neu24.7; 340879) from BD Biosciences, CCR7 (FITC; 150503;FAB197F-100) fromR&DSystems, and EGFR (PE; AY13; 352904)from BioLegend. We detected the expression of the EGFR.CARor amRs using anti-FLAG mAb (APC; L5; 637308). An anti-idiotype mAb was used to detect the expression of the CD19.CAR as previously described (21). All samples were acquiredon a BD LSRFortessa, and a minimum of 10,000 events wereacquired per sample. Samples were analyzed on FlowJo 9(FlowJo LLC).

    Western blot analysisT-cell lysateswere resuspended in 2� Laemelli buffer (Bio-Rad)

    in reducing or nonreducing conditions. To assess signalingthrough the CAR or amR, T cells on ice were incubated with 1 mgof anti-FLAG Ab (clone M2) for 15minutes and then 1 mg of goatanti-mouse secondary Ab (BD Biosciences) for an additional15 minutes. Cells were then transferred to a 37�C water bathfor the indicated time points and lysed with 4� Laemelli buffer.All lysates were separated in 4% to 15% SDS-PAGE gelsand transferred to polyvinylidene difluoride membranes (allBio-Rad). Blots were probed for human CD3z (Santa Cruz Bio-technology), p-Y142 CD3z (Abcam), pan-ERK (BD Biosciences),and pan-Akt, p-S473 Akt, and p-T202/Y204 MAPK (all CellSignaling Technology) diluted 1:1,000 in TBS-Tween/5% skimmilk.Membranes were then incubated withHRP-conjugated goatanti-mouse or goat anti-rabbit IgG (both Santa Cruz) at a dilutionof 1:3,000 and imagedusing the ECLSubstrate Kit on aChemiDocMP System (both Bio-Rad) according to the manufacturer'sinstructions.

    In vitro activationBiotinylated recombinant EGFR and EGFRvIII protein (Acro-

    Biosystems) were added to 96-well plates coated with 1 mg ofavidin (Thermo Fisher Scientific) at a 3:1 ratio. RecombinantEGFR-Fc and HER2-Fc (R&D Systems) were coated on 96-wellplates overnight at a concentration of 1 mg/well. T cells wereseeded in duplicate or triplicate for 6 hours, and supernatant wascollected for IFNg and T cells were assessed for CD69 by flowcytometry.

    Proliferation assayT cells were labeled with 1.5 mmol/L carboxyfluorescein dia-

    cetate succinimidyl ester (CFSE; Invitrogen) and plated withirradiated AsPC-1 at 4:1 effector-to-target (E:T) ratio in theabsence of exogenous cytokines. CFSE dilution of CAR-T cells oramR-T cells was analyzed on day 5 using flow cytometry (21). Theproliferation index was quantified using FlowJo 9.

    Long-term in vitro cytotoxicityTumor cells were seeded at 2.5–5 � 105 per well in 24-well

    plates. Donor-matched T cells normalized for transduction effi-ciency were added at 1:5 E:T ratio. On day 3–5 of coculture, cellswere collected, and the frequency of T cells and residual tumorscells wasmeasured by flow cytometry. Tumor cells were identifiedas CD19þ for BV173 tumor cells or CD4– CD8– in the case of alladherent tumor cell lines (27).

    Repetitive coculture assayFor multiple rounds of coculture, tumor cells were seeded at

    5 � 105 per well in 24-well plates. Donor-matched T cellsnormalized for transduction efficiency were added at 1:2 E:Tratio. On day 3 of coculture, a fraction of the cells wascollected, and the frequency of T cells and residual tumorscells was measured by flow cytometry. Between cocultures,T cells were washed and resuspended in fresh medium, with-out the addition of exogenous cytokines, and left to rest for 3days (27).

    Cytokine analysisSupernatant was collected from 0.5–1 � 105 CAR-T cells or

    amR-T cells plated in in vitro cytotoxicity assays at 1:5 E:T ratio after24 hours or from 1.25� 105 CAR-T cells or amR-T cells plated for6 hours. IFNg and IL2 were measured by ELISA per the manu-facturer's instructions (R&D Systems) in duplicate.

    Xenograft murine modelsSix- to 8-week-old male or female nonobese diabetic severe

    combined immunodeficiency/gc�/� (NSG) mice were injectedintravenously (i.v.) by tail vein with the Panc-1 tumor cell line(1� 106 cells/mouse) transducedwith theGFP-FFLuc reporter i.v.by tail vein injection. In other experiments, HPAF-II GFP-FFLuctagged cells were suspended in Matrigel and inoculated intraper-itoneally (i.p.; 1 � 106 cells/mouse). In rechallenge experiments,Panc-1 GFP-FFLuc cells (1 � 106 cells/mouse) were injected10 days prior to T-cell injection. Upon clearance of the Panc-1tumor cells, mice were then infused with BV173-HER2 cells (2 �106 cells/mouse). Mice were matched based on the biolumines-cence intensity and injected with 5� 106 or 1� 107 T cells i.v. 12to 14 days after tumor cell engraftment. The IVIS-Kinetic OpticalSystem (PerkinElmer) was used to monitor tumor burden. Micewere monitored and euthanized according to UNC-IACUCStandards.

    Statistical analysisData are reported as the mean and standard deviation, unless

    otherwise reported. To compare significant differences betweentwo samples, a two-tailed Student t test was applied. An ANOVAwith Tukey post hoc analysis was applied when a comparisonbetweenmultiple groups was required. A P value of less than 0.05was considered statistically significant. All figures were generatedusing GraphPad Prism (GraphPad Software).

    Bispecific Antibody Mimic Receptors

    www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 775

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • ResultsA soluble single-domain antibody targets EGFR in a bispecificmanner

    To develop a single-domain–based tumor antigen–bindingmoiety that targets two different epitopes of EGFR, we assembledthe previously identifiedZdomain–based EGFR-binding affibodyZEGFR:1907 and an FN3-based EGFR-binding monobody (17).The length and flexibility of the linker between the two EGFR-binding domains were optimized to retain the target-bindingaffinity, specificity, and independent folding of each EGFR-binding domain. We observed that when the linker is too short,both domains failed to bind targets. An overly long linker resultedin the loss of the bivalent effect as well as instability of the ligands.Because the linker length appeared to be antigen and binderdependent, we only used the linker length that worked well inthis proof-of-concept work. In this study, the two antigen-bindingmoieties were separated by a 25-residue flexible linker. Theresulting bispecific affinity molecule, from now on referred toas Bi-EGFR, binds to the extracellular domain of human EGFRat two nonoverlapping epitopes with an affinity (Kd) around0.38 � 0.07 nmol/L (Fig. 1A), which is approximately 10 timeshigher than that of the monomeric EGFR-binding FN3 domain(FN3.EGFR: 3.1 � 0.9 nmol/L; Fig. 1B) or Z domain (AFF.EGFR:3.2 � 0.3 nmol/L; Fig. 1C).

    Bi-EGFR.amR T cells resemble those expressing conventionalEGFR-specific CAR

    To construct the CAR vector, the sequence of the biochemicallyoptimized Bi-EGFR was fused to the CD28 and CD3z-chain sig-naling domains via the CD8a hinge and transmembrane domains(21). To detect the expression of the Bi-EGFR.amR in T cells (Bi-EGFR.amR-T cells) following transduction, we included a FLAG taginto the Bi-EGFR.amR cassette (Supplementary Fig. S1). A conven-tional EGFR-specific CAR (EGFR.CAR) generated using the scFvderived from themonoclonal antibody cetuximab (SupplementaryFig. S1), and theCD19-specific CAR (CD19.CAR; ref. 21)were usedas controls. AmRs composed of either the EGFR-A binding moiety(FN3.EGFR.amR) or the EGFR-B binding moiety (AFF.EGFR.amR)alone were also constructed (Supplementary Fig. S1). Upon retro-viral gene transfer, T cells stably expressed the Bi-EGFR.amR(Fig. 1D and E), expanded in vitro in response to exogenouscytokines (Fig. 1F), andmaintained T-cell composition comparablewith EGFR.CAR-T cells and CD19.CAR-T cells (Fig. 1G). Westernblot analysis of lysates from Bi-EGFR.amR-T cells detecting theCD3z chain under reducing conditions showed the native z-chain(17 kDa) and a band at the expected size of 55 kDa, indicating theintegrity of the assembled Bi-EGFR.amR (Fig. 1H). We then ana-lyzed proximal and distal signaling in EGFR.CAR-T cells and Bi-EGFR.amR-T cells upon receptor cross-linking. As shown in Fig. 1I,receptor cross-linking in both Bi-EGFR.amR-T cells and EGFR.CAR-T cells triggered similar phosphorylation of proximal (CD3z) anddistal (Akt and ERK) signaling molecules.

    Bi-EGFR.amR-T cells demonstrate activity against tumor cellsexpressing EGFR

    To demonstrate that Bi-EGFR.amR-T cells specifically targetEGFR, we used the EGFR– tumor cell line BV173 (BV173-WT)and transduced it to express EGFR (BV173-EGFR)with a retroviralvector encoding the full-length human EGFR (Supplementary Fig.S2A). We then cocultured BV173-WT or BV173-EGFR cells with

    control nontransduced T cells (NTs), CD19.CAR-T cells, EGFR.CAR-T cells, and Bi-EGFR.amR-T cells. NTs did not eliminateeither BV173-WT or BV173-EGFR cells, CD19.CAR-T cells elim-inated both cell types (Supplementary Fig. S2B and S2C), andBi-EGFR.amR-T cells and EGFR.CAR-T cells only eliminatedBV173-EGFR cells (Supplementary Fig. S2B and S2C), indicatingthe antigen specificity of the redirected T cells. Antitumor activitywas then tested against tumor cell lines that physiologicallyexpress EGFR (Supplementary Fig. S2D). In coculture experi-ments, Bi-EGFR.amR-T cells and EGFR.CAR-T cells demonstratedsimilar antitumor activity in vitro (Fig. 2A) and released compa-rable amounts of IFNg (Fig. 2B) and IL2 (Fig. 2C). Using a CFSEdilution assay, we demonstrated the proliferation of Bi-EGFR.amR-T cells in response to EGFR-expressing targets (Fig. 2Dand E). Finally, we evaluated the antitumor activity of Bi-EGFR.amR-T cells in a metastatic model of EGFR-expressing pancreaticcancer in NSG mice (Fig. 2F). Bi-EGFR.amR-T cells and EGFR.CAR-T cells equally controlled human Panc-1 tumor cell growthas assessed by measurement of tumor bioluminescence intensity(Fig. 2G and H).

    Bi-EGFR.amR-T cells recognize two nonoverlapping epitopesof EGFR

    AmRs composed of either the FN3.EGFR.amRor the AFF.EGFR.amR alone were transduced in T cells (Supplementary Fig. S3A).Both AFF.EGFR.amR-T cells and FN3.EGFR.amR-T cells showedcomparable activity in vitro against tumor cells expressing the full-length EGFR (Supplementary Fig. S3B–S3F) and proliferated inresponse to EGFR-expressing targets (Supplementary Fig. S3G andS3H). To demonstrate the bispecific feature of the Bi-EGFR, weanalyzed the binding of Bi-EGFR and each of its monomericdomains using the recombinant extracellular domain of thewild-type EGFR (wtEGFR) and the EGFRvIII (EGFRvIII) mutant,a constitutively active and ligand-independent variant of EGFRwith deletions in exons 2 to 7. We found that Bi-EGFR and Zdomain–based AFF.EGFR bound both wtEGFR and EGFRvIII.However, FN3 domain–based FN3.EGFR bound to wtEGFR butnot EGFRvIII (Supplementary Fig. S4A), suggesting that it recog-nizes an antigen encoded in the 267 AA that are absent inEGFRvIII. To assess recognition of the two different EGFR epi-topes, we used the extracellular domain of wtEGFR and EGFRvIIImutant recombinant proteins. Control, AFF.EGFR.amR-T cells,FN3.EGFR.amR-T cells, and Bi-EGFR.amR-T cells were seeded intissue culture plates coated with either wtEGFR or EGFRvIII, andthe CD69 expression and IFNg release by T cells were measured.Both wtEGFR and EGFRvIII recombinant proteins activated AFF.EGFR.amR-T cells and BiEGFR.amR-T cells, whereas only thewtEGFR protein activated FN3.EGFR.amR-T cells (Fig. 3A–C),indicating that the epitope recognized by the FN3.EGFR bindingmoiety is either located in the 267 AA region deleted in EGFRvIII,or the mutation has altered the epitope accessibility due to thedeletion-induced conformational changes. In contrast, the AFF.EGFR binding moiety recognizes an epitope that is conservedbetweenEGFRandEGFRvIII (Fig. 3A–C). To ensure that bothAFF.EGFR and FN3.EGFR binding moieties can induce the activationof T cells when assembled into the Bi-EGFR.amR, we generatedan AFF.EGFR.amR mutant–binding moiety (mAFF.EGFR.amR)in which critical residues at the EGFR-binding alpha heliceswere mutated to alanine to reduce binding to EGFR. As shownin Supplementary Fig. S4B, mAFF.EGFR.amR was expressedin T cells, but mAFF.EGFR.amR-T cells did not eliminate

    Ahn et al.

    Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research776

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • Figure 1.

    Bi-EGFR.amR-T cells and conventional EGFR.CAR-T cells show comparable activity. A–C, Biolayer interferometry (BLI) was used to measure the affinity of themonomeric or heterodimeric single-domain antibody mimics binding to recombinant Fc-EGFR on the Fort�eBio Octet system. Biotinylated antibody mimics(100 nmol/L) were immobilized onto a streptavidin biosensor. Binding kinetics were measured against various concentrations of EGFR (0, 25, 50, and100 nmol/L). The Kdwas calculated based on kinetic fitting. D, Representative expression of the Bi-EGFR.amR, EGFR.CAR, or CD19.CAR in T cells as assessed byflow cytometry. Activated T cells were transduced with retroviral vectors encoding the Bi-EGFR.amR, EGFR.CAR, or CD19.CAR. With the exception of the CD19.CAR, all constructs were detected using an anti-FLAG Ab. The CD19.CAR was detected using an anti-idiotype Ab. Shaded and unshaded histograms indicatenontransduced and specific mAb, respectively. E, Summary of amR or CAR expression (n¼ 4). F, Expansion kinetics of Bi-EGFR.amR-T cells, EGFR.CAR-T cells,or CD19.CAR-T cells NT (n¼ 4); error bars denote SD. G, Phenotypic composition of Bi-EGFR.amR-T cells, EGFR.CAR-T cells or CD19.CAR-T cells 10 days aftertransduction (n¼ 4); error bars denote SD. H, Reducing immunoblots of Bi-EGFR.amR-T cell and EGFR.CAR-T cell lysates. Immunoblots were probed withanti-CD3z. Top and bottom represent detection of the receptors and endogenous z-chain, respectively. I, EGFR.CAR-T cells and Bi-EGFR.amR-T cells wereincubated at the indicated time points with the anti-FLAG Ab and cross-linked with a secondary Ab to induce the aggregation of the receptors. Cell lysates wereimmunoblotted to detect proximal (CD3z p-Y142) and distal (Akt p-S473 and ERK p-T202/204) phosphorylation events following receptor cross-linking. TotalCAR.CD3z or amR.CD3z and endogenous CD3zwere used as loading controls. Data are representative of 4 experiments.

    Bispecific Antibody Mimic Receptors

    www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 777

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • BV173-EGFR cells (Supplementary Fig. S4C and S4D), indicatingthat the mutations had abrogated binding to EGFR. However, Tcells expressing a Bi-EGFR.amR constructed with the FN3.EGFRand mAFF.EGFR binding moieties (mBi-EGFR.amR) upregulatedCD69 and released IFNg when seeded in wells coated withwtEGFR recombinant protein, but not in response to EGFRvIIIprotein, indicating that the FN3.EGFR binding moiety alone caninduce T-cell activation when the function of the other EGFR-binding moiety (AFF.EGFR) is abolished in the mBi-EGFR.amR(Fig. 3D–F).

    Bispecific amR against HER2 demonstrates antitumor activityWe also constructed an amR receptor against another member

    of the ErbB family of receptor tyrosine kinase, HER2, to demon-strate the applicability of this technology to other TAAs. Using asimilar approach to the construction of Bi-EGFR, a tumorantigen–binding moiety that targets two different epitopes ofHER2 (Bi-HER2) was generated, which showed a HER2-binding affinity (Kd) around 0.25 � 0.04 nmol/L (Fig. 4A),compared with 1.8 � 0.6 nmol/L (Fig. 4B) of a HER2-bindingDARPin (15) and 1.3� 0.4 nmol/L of a HER2-binding Z domain

    Figure 2.

    Bi-EGFR.amR-T cells show activity against EGFR-expressing tumor cells in vitro and in vivo.A, Control T cells (NTs or CD19.CAR-T cells), Bi-EGFR.amR-T cells,and EGFR.CAR-T cells were cocultured with MCF-7 cells (EGFR negative) or EGFR-expressing pancreatic adenocarcinoma cell lines (AsPC-1, BxPC-3, HPAF-II,and Panc-1) at 1:5 E:T ratio. Cells were collected and quantified by flow cytometry on day 5. The frequency of residual tumor cells was identified as CD4�CD8� livecells (n¼ 3–4), P < 0.01 when Bi-EGFR.amR-T cells or EGFR.CAR-T cells are compared with control T cells; two-way ANOVAwith Tukey correction. B, IFNg and(C) IL2 released in the coculture supernatant by Bi-EGFR.amR-T cells, EGFR.CAR-T cells, and control T cells after 24 hours of coculture with tumor cells asassessed by ELISA (n¼ 3–4), P < 0.01 when Bi-EGFR.amR-T cells or EGFR.CAR-T cells are compared with control T cells; two-way ANOVAwith Tukey correction.D, Bi-EGFR.amR-T cells, EGFR.CAR-T cells or control T cells were labeled with CFSE and stimulated with irradiated EGFR-expressing AsPC-1 cells at 4:1 E:T.Representative CFSE dilution on day 5. E, Proliferation index as assessed by CFSE dilution (n¼ 4), P < 0.01 when Bi-EGFR.amR-T cells or EGFR.CAR-T cells arecompared with control T cells; one-way ANOVAwith Tukey correction. F, Schematic representation of a metastatic pancreatic cancer model in NSGmice usingthe FFLuc-labeled human Panc-1 cell line. Representative images of tumor bioluminescence (BLI) (G) and kinetics (H) of tumor growth as assessed by BLImeasurements. Data are representative of two independent experiments with 5 mice per group; P < 0.01 when Bi-EGFR.amR-T cells or EGFR.CAR-T cells arecompared with control T cells; two-way ANOVAwith Tukey correction.

    Ahn et al.

    Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research778

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • (ref. 18; Fig. 4C), respectively. Basedon thisHER2-binding ligand,a bispecific Bi-HER2.amR was constructed (Supplementary Fig.S5A). T cells stably expressed the Bi-HER2.amR (Bi-HER2.amR-Tcells) upon retroviral transfer, expanded in vitro in response toexogenous cytokines (Supplementary Fig. S5B) and maintainedT-cell composition comparable to control T cells (SupplementaryFig. S5C). Bi-HER2.amR-T cells demonstrated targeting of HER2-expressing cells (Supplementary Fig. S5D–S5F). Furthermore,Bi-HER2.amR-T cells exhibited antitumor activity (Fig. 4D) andreleased IFNg (Fig. 4E) and IL2 (Fig. 4F) in a HER2-dependentmanner when cocultured with tumor cell lines expressing HER2,but not with HER2-negative cell lines (Supplementary Fig. S5D).We also evaluated the efficacy of Bi-HER2.amR-T cells in a

    metastatic tumor model of HER2-expressing human HPAF-IIpancreatic cancer in NSG mice (Fig. 4G). Bi-HER2.amR-T cellsand EGFR.CAR-T cells equally controlled tumor cell growth asassessed by measurement of tumor bioluminescence intensity(Fig. 4H and I).

    Bispecific EGFR-HER2.amR-T cells target tumor cells expressingEGFR and HER2

    To generate amRs targeting two different TAAs, we integratedthe EGFR-specific FN3 binding moiety and the HER2-specificDARPin binding moiety and created the EGFR-HER2.amR witha 27-residue flexible linker between the two antigen-bindingmoieties (Supplementary Fig. S6A). T cells stably expressed the

    Figure 3.

    Bi-EGFR.amR-T cells are activated by recognition of two nonoverlapping epitopes on EGFR. Control (NTs), AFF.EGFR.amR-T cells, FN3.EGFR.amR-T cells, andBi-EGFR.amR-T cells were seeded in tissue culture plates coated with either recombinant human EGFRWT protein (EGFR) or the truncated mutant EGFRvIIIrecombinant protein (EGFRvIII).A, Representative expression of CD69 on NTs, AFF.EGFR.amR-T cells, FN3.EGFR.amR-T cells, and Bi-EGFR.amR-T cells 6 hoursafter stimulation with plate-bound EGFR or EGFRvIII protein as assessed by flow cytometry. Shaded and dashed histograms indicate media and PMA/Ionocontrols, respectively. Solid and dashed lines indicate EGFR and EGFRvIII protein stimulation, respectively. B, Summary of CD69 expression on total live T cells (n¼ 4), P < 0.01 when comparing FN3.EGFR.amR-T cells seeded in EGFR and EGFRvIII-coated wells; two-way ANOVAwith Tukey correction. C, IFNg released in thesupernatant by AFF.EGFR.amR-T cells, FN3.EGFR.amR-T cells, and Bi-EGFR.amR-T cells as assessed by ELISA (n¼ 3), P < 0.01 when comparing FN3.EGFR.amR-T cells seeded in rEGFR and rEGFRvIII-coated wells; two-way ANOVAwith Tukey correction. D,mAFF.EGFR.amR-T cells or mBi-EGFR.amR-T cells werecocultured with BV173-WT or BV173-EGFR cells at 1:5 E:T for 3 days. CD19.CAR-T cells were used as a positive control. Cells were collected, and T cells (CD3þ)and tumor cells (CD19þ) were quantified by flow cytometry. Representative flow plots are illustrated. E,Quantification of BV173-WT or BV173-EGFR cellsremaining after 3 days of coculture. F, IFNg released in the supernatant by NT, CD19.CAR-T, mAFF.EGFR.amR-T, and mBi-EGFR.amR-T cells as assessed by ELISA(n¼ 2–4), P < 0.01 when comparing the percentage of residual BV173-WT and BV173-EGFR cells remaining in the mBi-EGFR.amR-T wells; two-way ANOVAwithTukey correction.

    Bispecific Antibody Mimic Receptors

    www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 779

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • EGFR-HER2.amR (Supplementary Fig. S6B and S6C), expandedin vitro in response to exogenous cytokines (SupplementaryFig. S6D) and maintained T-cell composition comparable tomonospecific FN3.EGFR.amR-T cells and DARPin.HER2.amR-Tcells (Supplementary Fig. S6E). To establish the bispecificity ofEGFR-HER2.amR-T cells, we seeded control, FN3.EGFR.amR-Tcells, DARPin.HER2.amR-T cells, and EGFR-HER2.amR-T cells in

    tissue cultures plates coated with either the extracellular domainof rhEGFR or rhHER2 recombinant proteins and assessed CD69expression and IFNg release by T cells. EGFR-HER2.amR-T cellsexpressed CD69 (Supplementary Fig. S6F) and secreted IFNg(Supplementary Fig. S6G) when seeded in wells coated witheither rhEGFR or rhHER2 proteins, whereas FN3.EGFR.amR-Tcells and DARPin.HER2.amR-T cells were only activated by

    Figure 4.

    Bi-HER2.amR-T cells have antitumor activity against HER2-expressing primary cells in vitro and in vivo. A–C, Biolayer interferometry (BLI) was used to measurethe affinity of the monomeric or heterodimeric single-domain antibody mimics binding to recombinant Fc-HER2 using the Fort�eBio Octet system. Biotinylatedantibody mimics (100 nmol/L) were immobilized onto a streptavidin biosensor. Binding kinetics was measured against various concentrations of HER2 (0, 25, 50,100 nmol/L). The Kdwas calculated based on kinetic fitting. D, Control T (NT), Bi-HER2.amR-T cells, and EGFR.CAR-T cells were cocultured with Panc-1 cells (HERnegative) or HER2-expressing pancreatic adenocarcinoma cell lines (AsPC-1 and HPAF-II) at 1:5 E:T. Cells were collected and quantified by flow cytometry on day5. The frequency of residual tumor cells was identified as CD4� CD8� live cells (n¼ 3–6), P < 0.01 when comparing Bi-HER2.amR-T cells or EGFR.CAR-T cells withNTs; two-way ANOVAwith Tukey correction. E, IFNg and (F) IL2 released in the coculture supernatant by NTs, Bi-HER2.amR-T cells or EGFR.CAR-T cells after 24hours of coculture with tumor cells as assessed by ELISA (n¼ 3–6), P < 0.01 when comparing Bi-HER2.amR-T cells or EGFR.CAR-T cells with NTs; two-wayANOVAwith Tukey correction. G, Schematic representation of a metastatic pancreatic cancer model in NSGmice using the FFLuc-labeled human HPAF-II cellline. Representative images of tumor bioluminescence (BLI) (H) and kinetics (I) of tumor growth as assessed by BLI measurements. Data are representative oftwo independent experiments with 5 mice per group; P < 0.01 when Bi-HER2.amR-T cells or EGFR.CAR-T cells are compared with NTs; two-way ANOVAwithTukey correction.

    Ahn et al.

    Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research780

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • Figure 5.

    EGFR-HER2.amR-T cells show dual specificity. Coculture experiment in which T cells were plated with its respective BV173 target cells, and 3 days later T cellswere collected and replated with the same BV173 tumor cell line or BV173 cells expressing the nonspecific target. A, Schema of the repetitive cocultureexperiments with control (CD19.CAR-T cells), monospecific FN3.EGFR.amR-T cells, DARPin.HER2.amR-T cells, and bispecific EGFR-HER2.amR-T cells all plated at1:2 E:T ratio. B, Representative flow plots of the repetitive coculture experiments on the second round. Cells were collected and quantified by flow cytometry onday 3. The frequency of residual tumor cells was identified as CD3�CD19þ live cells. C,Quantification of tumor cells remaining after the second coculture (n¼ 2–3), P < 0.01 when comparing FN3.EGFR.amR-T cells and DARPin.HER2.amR-T cells and their respective targets againstWT tumor cells; two-way ANOVAwithTukey correction. D, IFNg released in the coculture supernatant by CD19.CAR-T cells, FN3.EGFR.amR-T cells, DARPin.HER2-amR-T cells, or EGFR-HER2.CAR-Tcells after 24 hours of coculture with tumor cells as assessed by ELISA (n¼ 2–3), P < 0.01 when comparing FN3.EGFR.amR-T cells and DARPin.HER2.amR-T cellsand their respective targets against WT cells; two-way ANOVAwith Tukey correction. E, Schematic representation of the rechallenge tumor model in NSGmiceusing the EGFRþHER2� human Panc-1 cell line labeled with the FF-Luc and the EGFR�HER2 human BV173-HER2 cell line. Representative images of the Pan-1tumor BLI (F) and BLI kinetics (G) of tumor growth of one experiment 3–7mice per group. H, Kaplan–Meier survival curve of NSGmice after rechallenge with theBV173-HER2 cell line; P < 0.01 when mice treated with EGFR.amR-T cells are compared with mice treated with EGFR-HER2.amR-T cells.

    Bispecific Antibody Mimic Receptors

    www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 781

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • rhEGFR and rhHER2, respectively. Next, we evaluated the func-tionality of EGFR-HER2.amR-T cells against BV173-WT, BV173-EGFR, and BV173-HER2 cells (Supplementary Fig. S6H). Weperformed a series of cocultures in which T cells were plated withrespective BV173 target cells, and 3 days later, T cells werecollected and replated with the same BV173 tumor cell line orBV173 cells expressing a nonspecific target (Fig. 5A). As shownin Fig. 5B–D, FN3.EGFR.amR-T cells and DARPin.HER2.amRcontinued to eliminate only BV173-EGFR and BV173-HER2,respectively, whereas EGFR-HER2.amR-T cells eliminated bothBV173-EGFR and BV173-HER2 targets in both the primary andsecondary cocultures. In order to demonstrate the bispecificity ofthe EGFR-HER2.amR-T cells in vivo, mice were engrafted with theEGFRþHER2� human Panc-1 cell line labeled with FF-Luc, andinfused with either EGFR.amR-T cells or EGFR-HER2.amR-T cells.Tumor growth was equally controlled by both EGFR.amR-T cellsand EGFR-HER2.amR-T cells (Fig. 5E–G). However, when thesemice were rechallenged with the EGFR�HER2þ BV173 tumor cellline, EGFR.amR-T-treated mice developed limb paresis due togrowth of the BV173 tumorwithin the spinal cord, whereas EGFR-HER2.amR-T cell–treated mice remained healthy (Fig. 5H).

    DiscussionThe generation ofmultiredirected CAR-T cellsmay be necessary

    to effectively eradicate tumors inwhich TAAsof interest canbe lostor are heterogeneously expressed in tumor cells. Here, we dem-onstrated that modular single-domain antibody mimics are apractical alternative to the conventional scFvs to generate T cellswith engineered multiple antigen-targeting features. We con-structed antibody mimics that can be assembled with signalingmolecules of the T-cell receptor and costimulatory endodomains.We further demonstrated the functionality in vitro and in vivo ofT cells expressing antibody mimics showing their ability torecognize simultaneously two epitopes of the same antigens ortwo distinct antigens.

    In this proof-of-principle study, we have assessed whethersingle-domain antibody mimics can be used to redirect thespecificity of human T lymphocytes (30, 31). We integratedsingle-domain antibody mimics binding to the nonoverlappingregions of EGFR to create an EGFR-binding ligand with bispeci-ficity. We showed that T cells expressing the single-domain AFF.EGFR.amR or FN3.EGFR.amR have comparable activity in vitroand in vivo to the conventional scFv-based cetuximab EGFR.CAR.To further demonstrate the applicability of redirecting T-cellspecificity by using antibody mimics, we also validated theapproach by targeting HER2 expressing tumors using a Bi-HER2targeting ligand.

    Targeting two epitopes of the samemolecule may help preventtumor escape when the targeted antigen can be expressed by

    alternative mRNA splicing, which may cause loss of a targetedepitope (6). When the AFF.EGFR and FN3.EGFR were combinedin one single amR cassette, we demonstrated that two epitopes ofEGFR can be targeted without causing detrimental effects in Tcells. Targeting two distinct antigens expressed by tumor cellsremains challenging. We demonstrated that single-domain anti-bodymimics can be assembled in one single cassette to efficientlytarget two different antigens. As a proof of principle, we showedthat both EGFR and HER2 can be effectively targeted by dual-specific amRs neither with impairing targeting of each singleantigen nor with detrimental effects of engineered T cells.

    In summary, we provided proof of concept that antibodymimics can be used to generate combinatorial targeting of engi-neered T cells. Taking in consideration that antibody mimics aregenerated synthetically, our proposed approach can be adapted toa systematic screening of combinatorial antigens to be tested inhuman malignancies.

    Disclosure of Potential Conflicts of InterestR. Liu is the founder of, has ownership interest in, and is a consultant/

    advisory board for Panacise Bio Inc. No potential conflicts of interest weredisclosed by the other authors.

    Authors' ContributionsConception and design: S. Ahn, J. Li, B. Savoldo, R. Liu, G. DottiDevelopment of methodology: S. Ahn, J. Li, R. Liu, G. DottiAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): S. Ahn, J. Li, C. Sun, K. Gao, K. Hirabayashi, H. LiAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): S. Ahn, J. Li, C. Sun, K. Gao, R. Liu, G. DottiWriting, review, and/or revision of the manuscript: S. Ahn, J. Li, K. Gao,B. Savoldo, R. Liu, G. DottiAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): S. Ahn, R. LiuStudy supervision: B. Savoldo, R. Liu, G. Dotti

    AcknowledgmentsThis work was partially supported by the R01CA157738 from the NCI to

    R. Liu, University Cancer Research Fund (UCRF) to G. Dotti, R01-CA193140-03from the NCI to G. Dotti, and innovation grants from the Eshelman Institutefor Innovation RX03612118 and RX03712112 to R. Liu. The UNC ImagingCore is supported in part by an NCI core grant (P30-CA016086-40). The UNCFlow Cytometry Core is supported in part by the North Carolina BiotechCenter Institutional Support grant (2012-IDG-1006). We thank Dr. AshutoshTripathy for assistance in the biophysical analyses of the bispecific antigenreceptors.

    The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

    Received September 11, 2018; revised December 6, 2018; accepted February27, 2019; published first March 6, 2019.

    References1. Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and

    targeting of cytotoxic lymphocytes through chimeric single chainsconsisting of antibody-binding domains and the gamma or zeta sub-units of the immunoglobulin and T-cell receptors. Proc Natl Acad SciUSA 1993;90:720–4.

    2. Dotti G, Gottschalk S, Savoldo B, Brenner MK. Design and development oftherapies using chimeric antigen receptor-expressing T cells. Immunol Rev2014;257:107–26.

    3. Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigenreceptor design. Cancer Discov 2013;3:388–98.

    4. Maude SL, FreyN, ShawPA, Aplenc R, Barrett DM, BuninNJ, et al. Chimericantigen receptor T cells for sustained remissions in leukemia. N Engl J Med2014;371:1507–17.

    5. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO,Stetler-Stevenson M, et al. Chemotherapy-refractory diffuse large B-celllymphoma and indolent B-cell malignancies can be effectively treated with

    Cancer Immunol Res; 7(5) May 2019 Cancer Immunology Research782

    Ahn et al.

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • autologous T cells expressing an anti-CD19 chimeric antigen receptor.J Clin Oncol 2015;33:540–9.

    6. Sotillo E, Barrett DM, Black KL, Bagashev A, Oldridge D, Wu G, et al.Convergence of acquired mutations and alternative splicing of CD19enables resistance to CART-19 immunotherapy. Cancer Discov 2015;5:1282–95.

    7. O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morris-sette JJD, et al. A single dose of peripherally infused EGFRvIII-directed CART cells mediates antigen loss and induces adaptive resistance in patientswith recurrent glioblastoma. Sci Transl Med 2017;9.

    8. Grada Z,HegdeM, Byrd T, ShafferDR,Ghazi A, Brawley VS, et al. TanCAR: anovel bispecific chimeric antigen receptor for cancer immunotherapy.Mol Ther Nucleic Acids 2013;2:e105.

    9. Hegde M, Mukherjee M, Grada Z, Pignata A, Landi D, Navai SA, et al.Tandem CAR T cells targeting HER2 and IL13Ralpha2 mitigate tumorantigen escape. J Clin Invest 2016;126:3036–52.

    10. Cotten SW, Zou J, Valencia CA, Liu R. Selection of proteins with desiredproperties from natural proteome libraries using mRNA display.Nat Protoc 2011;6:1163–82.

    11. Koide A, Bailey CW,Huang X, Koide S. The fibronectin type III domain as ascaffold for novel binding proteins. J Mol Biol 1998;284:1141–51.

    12. Nygren PA. Alternative binding proteins: affibody binding proteins devel-oped from a small three-helix bundle scaffold. FEBS J 2008;275:2668–76.

    13. BinzHK,Amstutz P, PluckthunA. Engineeringnovel binding proteins fromnonimmunoglobulin domains. Nat Biotechnol 2005;23:1257–68.

    14. Binz HK, Pluckthun A.Engineered proteins as specific binding reagents.Curr Opin Biotechnol 2005;16:459–69.

    15. Zahnd C, Pecorari F, Straumann N, Wyler E, Pluckthun A. Selection andcharacterization of Her2 binding-designed ankyrin repeat proteins. J BiolChem 2006;281:35167–75.

    16. Fellouse FA, Esaki K, Birtalan S, Raptis D, Cancasci VJ, Koide A, et al. High-throughput generation of synthetic antibodies from highly functionalminimalist phage-displayed libraries. J Mol Biol 2007;373:924–40.

    17. Friedman M, Orlova A, Johansson E, Eriksson TL, H€oid�en-Guthenberg I,Tolmachev V, et al. Directed evolution to low nanomolar affinity of atumor-targeting epidermal growth factor receptor-binding affibody mol-ecule. J Mol Biol 2008;376:1388–402.

    18. Orlova A, Magnusson M, Eriksson TL, Nilsson M, Larsson B, H€oid�en-Guthenberg I, et al. Tumor imaging using a picomolar affinity HER2binding affibody molecule. Cancer Res 2006;66:4339–48.

    19. Steiner D, Forrer P, Pluckthun A. Efficient selection of DARPins with sub-nanomolar affinities using SRP phage display. J Mol Biol 2008;382:1211–27.

    20. Vazquez-Lombardi R, Phan TG, Zimmermann C, Lowe D, Jermutus L,Christ D. Challenges and opportunities for non-antibody scaffold drugs.Drug Discov Today 2015;20:1271–83.

    21. Diaconu I, Ballard B, Zhang M, Chen Y, West J, Dotti G, et al. Induciblecaspase-9 selectively modulates the toxicities of CD19-specific chimericantigen receptor-modified T cells. Mol Ther 2017;25:580–92.

    22. Dotti G, Savoldo B, Pule M, Straathof KC, Biagi E, Yvon E, et al. Humancytotoxic T lymphocytes with reduced sensitivity to Fas-induced apoptosis.Blood 2005;105:4677–84.

    23. KimD, FriedmanAD, LiuR. Tetraspecific ligand for tumor-targeteddeliveryof nanomaterials. Biomaterials 2014;35:6026–36.

    24. Ramos CA, Ballard B, Zhang H, Dakhova O, Gee AP, Mei Z, et al. Clinicaland immunological responses after CD30-specific chimeric antigen recep-tor-redirected lymphocytes. J Clin Invest 2017;127:3462–71.

    25. Ramos CA, Savoldo B, Torrano V, Ballard B, Zhang H, Dakhova O, et al.Clinical responses with T lymphocytes targeting malignancy-associatedkappa light chains. J Clin Invest 2016;126:2588–96.

    26. Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, et al. Closelyrelated T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 2014;123:3750–9.

    27. Hoyos V, Savoldo B, Quintarelli C,Mahendravada A, ZhangM, Vera J, et al.Engineering CD19-specific T lymphocytes with interleukin-15 and a sui-cide gene to enhance their anti-lymphoma/leukemia effects and safety.Leukemia 2010;24:1160–70.

    28. Vera JF, Hoyos V, Savoldo B, Quintarelli C, Giordano Attianese GM, LeenAM, et al. Genetic manipulation of tumor-specific cytotoxic T lymphocytesto restore responsiveness to IL-7. Mol Ther 2009;17:880–8.

    29. Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, et al. Heparanasepromotes tumor infiltration and antitumor activity of CAR-redirected Tlymphocytes. Nat Med 2015;21:524–9.

    30. Han X, Cinay GE, Zhao Y, Guo Y, Zhang X, Wang P. Adnectin-based designof chimeric antigen receptor for T cell engineering. Mol Ther 2017;25:2466–76.

    31. Siegler E, Li S, Kim YJ, Wang P. Designed ankyrin repeat proteins as Her2targeting domains in chimeric antigen receptor-engineered T cells.Hum Gene Ther 2017;28:726–36.

    www.aacrjournals.org Cancer Immunol Res; 7(5) May 2019 783

    Bispecific Antibody Mimic Receptors

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/

  • 2019;7:773-783. Published OnlineFirst March 6, 2019.Cancer Immunol Res Sarah Ahn, Jingjing Li, Chuang Sun, et al. That Mimic AntibodiesCancer Immunotherapy with T Cells Carrying Bispecific Receptors

    Updated version

    10.1158/2326-6066.CIR-18-0636doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://cancerimmunolres.aacrjournals.org/content/suppl/2019/03/06/2326-6066.CIR-18-0636.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://cancerimmunolres.aacrjournals.org/content/7/5/773.full#ref-list-1

    This article cites 30 articles, 8 of which you can access for free at:

    Citing articles

    http://cancerimmunolres.aacrjournals.org/content/7/5/773.full#related-urls

    This article has been cited by 3 HighWire-hosted articles. Access the articles at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected]

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://cancerimmunolres.aacrjournals.org/content/7/5/773To request permission to re-use all or part of this article, use this link

    on June 3, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

    Published OnlineFirst March 6, 2019; DOI: 10.1158/2326-6066.CIR-18-0636

    http://cancerimmunolres.aacrjournals.org/lookup/doi/10.1158/2326-6066.CIR-18-0636http://cancerimmunolres.aacrjournals.org/content/suppl/2019/03/06/2326-6066.CIR-18-0636.DC1http://cancerimmunolres.aacrjournals.org/content/7/5/773.full#ref-list-1http://cancerimmunolres.aacrjournals.org/content/7/5/773.full#related-urlshttp://cancerimmunolres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerimmunolres.aacrjournals.org/content/7/5/773http://cancerimmunolres.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice