broadening specificity and enhancing cytotoxicity of ... · research article broadening...

11
Research Article Broadening Specicity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy Damiana Antonia Fa e 1 , Debora Martorelli 1 , Katy Mastorci 1 , Elena Muraro 1 , Jessica Dal Col 1 , Giovanni Franchin 2 , Luigi Barzan 3 , Elisa Comaro 1 , Emanuela Vaccher 4 , Antonio Rosato 5 , and Riccardo Dolcetti 1 Abstract Although promising, clinical responses to adoptive immuno- therapy for nasopharyngeal carcinoma (NPC) are still limited by the restricted number of EpsteinBarr virus (EBV) antigens that can be targeted and their poor immunogenicity. Our previous work indicated that the immunogenic features of the NPC- associated viral antigen BARF1 may be exploited for immuno- therapeutic purposes. Nevertheless, T-cell lines obtained with current protocols include only negligible numbers of BARF1- specic cytotoxic T lymphocytes, pointing to the need to enrich these effectors in BARF1 specicities. Considering that in B lymphocytes BARF1 is mainly a lytic EBV antigen, we tested different EBV lytic-cycle inducers (TPA/butyric acid, doxorubi- cin, and cisplatin) used at suboptimal concentrations for their ability to upregulate BARF1 expression in lymphoblastoid B-cell lines (LCL), the commonly used antigen-presenting cells, with- out compromising their survival. The LCLs treated with doxo- rubicin (DX-LCL) can reproducibly and efciently generate EBV-specic effectors enriched in BARF1 specicities from both healthy donors and NPC patients. These DX-LCLs also had more pronounced immunogenic properties, including HLA class I upregulation and expression of immunogenic cell death markers, such as enhanced calreticulin exposure and HMGB1 release. In particular, doxorubicin triggers an HMGB1 autocrine/ paracrine loop with its receptor, TLR4, which is also upregulated in DX-LCLs and is responsible for NF-kB activation and a delayed apoptosis that allows a prolonged stimulation of EBV-specic T-cell precursors. This protocol may thus constitute a valid alternative to the use of engineered LCLs to generate EBV-specic T-cell lines for adoptive immunotherapy, being relatively simple, easily upgradable to Good Manufacturing Practice standards, and therefore more broadly applicable. Cancer Immunol Res; 4(5); 43140. Ó2016 AACR. Introduction Nasopharyngeal carcinoma (NPC) is a malignant tumor arising from the mucosal epithelium of the nasopharynx, which is characterized by a multifactorial etiology and a peculiar geograph- ical distribution. In fact, NPC is relatively rare in most parts of the world, particularly in Europe and North America, while it has a high incidence in southern China and southeast Asia, where it constitutes a signicant health problem (1, 2). NPC is character- ized by several histopathologic entities, with the undifferentiated form being the most frequent (3, 4). The standard treatment for NPC is radiotherapy with or without chemotherapy, depending on disease stage. Although excellent control can be achieved with a combination of radiotherapy and chemotherapy, local recur- rence still occurs in 5% to 10% of cases (3, 4), and prognosis of patients with metastatic disease remains poor (3, 4). Hence, new low-toxicity therapeutic strategies are strongly needed to improve the clinical control of NPC. In addition to dietary, environmental, and genetic risk factors, this malignancy shows a strong association with EpsteinBarr virus (EBV), as supported by the observation that the EBV genome is monoclonal and detected in tumor cells of virtually all cases of NPC (5, 6). EBV infections in NPC cells are characterized by the so-called type II latency, with the expression of a limited set of latent proteins, including EBNA1, LMP-1, LMP-2, and BARF1, which may cooperatively contribute to EBV-driven epithelial cell transformation. These viral antigens are almost constantly, albeit heterogeneously, expressed by NPC cells and have become attrac- tive targets for immunotherapy. Adoptive infusion of autologous EBV-specic T cells has recently emerged as an effective tool for the treatment of EBV-driven malignancies, and its efcacy in lym- phoid tumors such as posttransplantation lymphoproliferative disease has been convincingly demonstrated (7). Phase I/II clin- ical studies have also provided evidence supporting the potential application of cytotoxic T lymphocyte (CTL)based therapy to NPC patients, either as a single treatment for recurrent or meta- static disease or in combination with chemotherapy (710). Nevertheless, the rate of clinical responses obtainable in NPC patients by adoptive immunotherapy is unsatisfactory. These disappointing ndings may be due to the limited set of viral antigens expressed by NPC cells, as well as to their poor immu- nogenicity, as shown by the subdominance of specic T-cell 1 Cancer Bio-Immunotherapy Unit, CRO-IRCCS, National Cancer Insti- tute, Aviano, Italy. 2 Division of Radiotherapy, CRO-IRCCS, National Cancer Institute, Aviano, Italy. 3 Otolaryngology, Azienda Ospedaliera S. Maria degli Angeli, Pordenone, Italy. 4 Medical Oncology Division A, CRO-IRCCS, National Cancer Institute, Aviano, Italy. 5 Istituto Oncolo- gico Veneto, IOV-IRCCS, Padua, Italy. Note: Supplementary data for this article are available at Cancer Immunology Research Online (http://cancerimmunolres.aacrjournals.org/). Corresponding Author: Riccardo Dolcetti, The University of Queensland Diamantina Institute, Brisbane, Australia. Phone: 61-7-34436953: Fax: 61-7- 34436966; E-mail: [email protected] doi: 10.1158/2326-6066.CIR-15-0108 Ó2016 American Association for Cancer Research. Cancer Immunology Research www.aacrjournals.org 431 on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Upload: others

Post on 26-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

Research Article

Broadening Specificity and EnhancingCytotoxicity of Adoptive T Cells forNasopharyngeal Carcinoma ImmunotherapyDamiana Antonia Fa�e1, Debora Martorelli1, Katy Mastorci1, Elena Muraro1,Jessica Dal Col1, Giovanni Franchin2, Luigi Barzan3, Elisa Comaro1,Emanuela Vaccher4, Antonio Rosato5, and Riccardo Dolcetti1

Abstract

Although promising, clinical responses to adoptive immuno-therapy for nasopharyngeal carcinoma (NPC) are still limited bythe restricted number of Epstein–Barr virus (EBV) antigens thatcan be targeted and their poor immunogenicity. Our previouswork indicated that the immunogenic features of the NPC-associated viral antigen BARF1 may be exploited for immuno-therapeutic purposes. Nevertheless, T-cell lines obtained withcurrent protocols include only negligible numbers of BARF1-specific cytotoxic T lymphocytes, pointing to the need to enrichthese effectors in BARF1 specificities. Considering that in Blymphocytes BARF1 is mainly a lytic EBV antigen, we testeddifferent EBV lytic-cycle inducers (TPA/butyric acid, doxorubi-cin, and cisplatin) used at suboptimal concentrations for theirability to upregulate BARF1 expression in lymphoblastoid B-celllines (LCL), the commonly used antigen-presenting cells, with-out compromising their survival. The LCLs treated with doxo-

rubicin (DX-LCL) can reproducibly and efficiently generateEBV-specific effectors enriched in BARF1 specificities from bothhealthy donors and NPC patients. These DX-LCLs also hadmore pronounced immunogenic properties, including HLAclass I upregulation and expression of immunogenic cell deathmarkers, such as enhanced calreticulin exposure and HMGB1release. In particular, doxorubicin triggers an HMGB1 autocrine/paracrine loop with its receptor, TLR4, which is also upregulatedin DX-LCLs and is responsible for NF-kB activation anda delayed apoptosis that allows a prolonged stimulation ofEBV-specific T-cell precursors. This protocol may thus constitutea valid alternative to the use of engineered LCLs to generateEBV-specific T-cell lines for adoptive immunotherapy, beingrelatively simple, easily upgradable to Good ManufacturingPractice standards, and therefore more broadly applicable.Cancer Immunol Res; 4(5); 431–40. �2016 AACR.

IntroductionNasopharyngeal carcinoma (NPC) is amalignant tumor arising

from the mucosal epithelium of the nasopharynx, which ischaracterized by amultifactorial etiology andapeculiar geograph-ical distribution. In fact, NPC is relatively rare in most parts of theworld, particularly in Europe and North America, while it has ahigh incidence in southern China and southeast Asia, where itconstitutes a significant health problem (1, 2). NPC is character-ized by several histopathologic entities, with the undifferentiatedform being the most frequent (3, 4). The standard treatment forNPC is radiotherapy with or without chemotherapy, dependingon disease stage. Although excellent control can be achieved witha combination of radiotherapy and chemotherapy, local recur-

rence still occurs in 5% to 10% of cases (3, 4), and prognosis ofpatients with metastatic disease remains poor (3, 4). Hence, newlow-toxicity therapeutic strategies are strongly needed to improvethe clinical control of NPC.

In addition to dietary, environmental, and genetic risk factors,this malignancy shows a strong association with Epstein–Barrvirus (EBV), as supported by the observation that the EBV genomeis monoclonal and detected in tumor cells of virtually all casesof NPC (5, 6). EBV infections in NPC cells are characterized bythe so-called type II latency, with the expression of a limited set oflatent proteins, including EBNA1, LMP-1, LMP-2, and BARF1,which may cooperatively contribute to EBV-driven epithelial celltransformation. These viral antigens are almost constantly, albeitheterogeneously, expressed by NPC cells and have become attrac-tive targets for immunotherapy. Adoptive infusion of autologousEBV-specific T cells has recently emerged as an effective tool for thetreatment of EBV-driven malignancies, and its efficacy in lym-phoid tumors such as posttransplantation lymphoproliferativedisease has been convincingly demonstrated (7). Phase I/II clin-ical studies have also provided evidence supporting the potentialapplication of cytotoxic T lymphocyte (CTL)–based therapy toNPC patients, either as a single treatment for recurrent or meta-static disease or in combination with chemotherapy (7–10).Nevertheless, the rate of clinical responses obtainable in NPCpatients by adoptive immunotherapy is unsatisfactory. Thesedisappointing findings may be due to the limited set of viralantigens expressed by NPC cells, as well as to their poor immu-nogenicity, as shown by the subdominance of specific T-cell

1Cancer Bio-Immunotherapy Unit, CRO-IRCCS, National Cancer Insti-tute, Aviano, Italy. 2Division of Radiotherapy, CRO-IRCCS, NationalCancer Institute, Aviano, Italy. 3Otolaryngology, Azienda OspedalieraS. Maria degli Angeli, Pordenone, Italy. 4Medical Oncology Division A,CRO-IRCCS, National Cancer Institute, Aviano, Italy. 5Istituto Oncolo-gico Veneto, IOV-IRCCS, Padua, Italy.

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

Corresponding Author: Riccardo Dolcetti, The University of QueenslandDiamantina Institute, Brisbane, Australia. Phone: 61-7-34436953: Fax: 61-7-34436966; E-mail: [email protected]

doi: 10.1158/2326-6066.CIR-15-0108

�2016 American Association for Cancer Research.

CancerImmunologyResearch

www.aacrjournals.org 431

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 2: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

responses usually elicited by LMP-1 and LMP-2 (11). Theselimitations are stimulating the exploration of novel strategiesto improve the efficacy of adoptive immunotherapy for NPC.Genetic manipulation of dendritic cells and lymphoblastoidcell lines (LCL), the conventional antigen-presenting cells (APC)used in current protocols, was recently shown to induce EBV-specific CTL cultures enriched in specificities for LMP antigensable to mediate enhanced clinical responses in patients withlymphoma (12); however, the applicability of such procedure islimited to very few specialized centers.

One of the possible strategies to overcome these limitations isthe exploitation of additional viral proteins expressed by NPCcells, whichmay serve as tumor-associated antigens to be targetedfor improved CTL induction and expansion. The oncogenic EBVprotein BARF1 is expressed in the majority of NPC cases (13–16)and may constitute an attractive therapeutic target. In particular,we have previously demonstrated that NPC patients show strongspontaneous CD4þ and CD8þ T-cell responses against BARF1protein and derived epitopes, indicating that BARF1 is a naturallyimmunogenic protein that may be targeted by specific CTLs (17).Indeed,wehave also shown that BARF1-specificCTLs canbe easilygenerated fromEBVþ donors, an important prerequisite to exploitBARF1 immunogenicity for immunotherapeutic purposes. Nev-ertheless, EBV-specific T-cell lines generated by current protocolsbased on the use of autologous EBVþ LCLs as APCs showonly verylimited specificities for BARF1, if any (17).

On these grounds,weperformed a study aiming at developing anew and broadly applicable protocol for adoptive immunother-apy of NPC, based on the generation of T-cell lines enriched inBARF1-specific effectors. Considering that in B lymphocytesBARF1 is mainly a lytic EBV antigen (18), we devised a strategybased on the use of low doses of EBV lytic cycle inducers toupregulate BARF1 in LCLs without inducing massive cell apopto-sis or a complete EBV lytic replication, to allow these cells toeffectively present BARF1 peptides together with other targetepitopes. To these ends, we investigated different EBV lytic cycleinducers, such as TPAwithbutyric acid, doxorubicin, and cisplatin(19, 20), which were used at suboptimal concentrations toupregulate BARF1 expression in LCLs without compromising cellsurvival. Our results demonstrate that LCLs treated with doxoru-bicin can reproducibly and efficiently generate EBV-specific effec-tors enriched in BARF1 specificities fromboth healthy donors andNPC patients. Moreover, doxorubicin-treated LCLs are alsoendowed with more pronounced immunogenic propertiesallowing the generation of EBV-specific CTLs with enhancedeffector functions. Our protocol may therefore constitute avalid alternative to the use of engineered LCLs to generateEBV-specific T-cell lines for adoptive immunotherapy, beingrelatively simple and designed to fully exploit the immuno-genic properties of doxorubicin, a drug largely used in theclinic. These characteristics make our protocol easily upgrad-able to Good Manufacturing Practice (GMP) standards andtherefore more broadly applicable.

Materials and MethodsCell lines and culture conditions

The following cell lines, all obtained in 2010 to 2012, wereused: c666.1 NPC (obtained from T. Ooka, Lyon; Granta-519mantle cell lymphoma (ACC-342, DMSZ); T2-A2 (kind gift fromM.G. Masucci, Stockholm); K562 (ACC-10, DMSZ); donor-

derived EBV-infected LCLs, generated by in vitro transformationof B cells with the B95-8 EBV strain. Short tandem repeat (STR)loci for cell lines authentication were evaluated for all cell lines inour laboratory in 2011 and at the end of the study in 2014, byusing the GenePrint10-System (Promega). All cell lines werecultured in RPMI-1640 (Gibco), containing 10% fetal bovineserum (Gibco), 2 mmol/L of L-glutamine, 100 mg/mL of strepto-mycin and 100 IU/mL of penicillin (Sigma), with the exception ofGranta-519, cultured in complete Dulbecco's Modified EagleMedium (Cambrex). Donor- and patient-derived BARF1-specificCTL lines were cultured in CellGro GMP DC (CellGenix), sup-plemented with 100 mg/mL of streptomycin and 100 IU/mL ofpenicillin (Sigma). LCLs were seeded in complete RPMI-1640medium (5 � 105 cells/mL) and treated with either 20 ng/mL of12-O-tetradecanoyl-phorbol-1-acetate (TPA) and 5 mmol/L ofsodium butyrate (NaB; Sigma) for 48 hours or doxorubicin,25 nmol/L for 6 hours and cisplatin, 5 mmol/L for 6 hours. Afterdoxorubicin and cisplatin treatment, cells were washed once andcultured in fresh complete medium for further 24 hours. Doxo-rubicin and cisplatin were provided by the anticancer drug unit ofour institution.

Generation of EBV-specific CTL linesDonor- and patient-derived BARF1-specific CTLs were gener-

ated and weekly restimulated using as APCs autologous LCLstreated or not with suboptimal concentrations of TPA þ NaB,doxorubicin, or cisplatin. After treatment, LCLs were g-irradiated80 Gy before the first stimulation, and 40 Gy before each otherrestimulation. IL2 (3 ng/mL) was added to the culture startingfrom day 14, and fresh medium was replaced every 3 days.Effectors were cocultured with APCs at a 40:1 T-cell:LCL ratio.

Multispectral imaging flow cytometryThe following fluorescent-conjugated monoclonal antibodies

were used: anti–CD8-PC7 (mouse IgG1; clone SFCI21Thy2D3,Beckman Coulter), anti–CD19-PE (mouse IgG1; clone HIB19,eBioscience), anti–granzyme-b–FITC (mouse IgG1; clone GB11)(BD Pharmingen). To determine the T:APC conjugate formation(CD8þ T cells and autologous B-LCL) and to enumerate gran-zyme-b granules, a cytotoxicity assay was performed. Briefly, after2 hours of coculture, autologous LCLþT cells (1.5 � 106 cells/condition) were stained with anti–CD8-PC7 and anti–CD19-PEantibodies in an appropriate volume of 10% rabbit serum andPBS. Then, cells were incubated with fixation/permeabilizationbuffer for 30minutes at 4�C,washed twice, and labeledwith anti–granzyme-b antibody in the presence of 2% rabbit serum in PBS at4�C for 45minutes and after twowashes cells were resuspended inPBSwith 1%paraformaldehyde. Cells were run on ImageStreamXcytometer using the INSPIRE software and images were analyzedusing the IDEAS software (Amnis). Cells were excited with a 488-nm laser (50 mW intensity). Brightfield, side scatter, and fluores-cent cell images were acquired at�40magnification. Only eventswith brightfield areas >30 mm2 (excluding debris) and nonsatu-rating pixels were collected. In T:APC binding experiments, 3 �104 events were collected for each sample. Cells were gated forfocused populations and doublets containing at least one T cellwere gated from among all cells. Intracellular granzyme-b gran-ules formation was determined by subcellular localization andspot count experiments. Events (3,000) were collected for eachsample. Cytoplasmic localization of the granules was measuredusing the IDEAS software "internalization algorithm," defined as

Fa�e et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology Research432

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 3: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

the ratio between intensity inside the cell and the intensity of theentire cell. The inside of the cell is defined by the "erosion mask"that fits the cell membrane. Cells containing small concentratedfluorescent spots have positive scores, whereas cells showinglittle/diffuse fluorescence have negative scores. Only viable cellswere selected on the basis of morphology. Single-stained controlswere used to compensate fluorescence between channel imageson a pixel-by-pixel basis.

For patients and donors, generation of HLA-A�0201-expres-sing c666.1 NPC cells, quantitative real-time PCR, immuno-blotting, cytotoxicity assays, HMGB1-release quantification,and TLR-4 neutralization experiments refer to SupplementaryMaterial.

ResultsDoxorubicin upregulates BARF1 mRNA expression

Different EBV lytic cycle inducers were used at suboptimalconcentrations to induce a mainly abortive lytic cycle and upre-

gulate BARF1 expression in LCLs without inducing major apo-ptotic effects. As a first step, we used the TPAþNaB combinationas the most commonly used treatment to trigger EBV lytic reac-tivation (2, 21). Healthy donor–derived LCLs treated for 48 hourswith TPA þ NaB showed significantly increased BARF1 mRNAexpression (2.5 mean fold-increase, P � 0.05; Fig. 1A). Neverthe-less, the treatment also induced a broad upregulation of bothlatent (LMP-1, EBNA-1, EBNA-2) and lytic (ZEBRA, EA) EBVgenes, together with a highly productive lytic cycle (Fig. 1A;Supplementary Table S1 and Fig. S1A), which was invariablyassociated withmarked levels of LCL apoptosis (Fig. 1B). A strongEBV reactivation was also induced by cisplatin, whereas doxoru-bicin-treated LCLs (DX-LCLs) showed amarked increase inBARF1mRNA expression levels and a lower, but still significant (P �0.05), upregulation of other EBV genes without inducing a mas-sive EBV lytic replication (Fig. 1A; Supplementary Fig. S1A).Notably, doxorubicin had less pronounced proapoptotic effectsas compared with the other drugs investigated, as shown by PARPcleavage analysis (Fig. 1B).

Figure 1.A, doxorubicin (DX) enhances BARF1 expression. qRT-PCR for EBV lytic and latent gene expression performed on LCLs treated with TPA þ NaB, doxorubicin,or cisplatin (CSP). mRNA fold expression was normalized relative to control LCLs (Ctrl-LCLs). Mean of three independent experiments. � , P � 0.05statistical significance for fold increase of BARF1 mRNA expression versus other EBV genes. �� , P � 0.05 denotes statistical significance of fold increase versusCtrl-LCLs (Student t test). B, DX-LCLs show a limited late apoptosis. PARP cleavage was assessed at the end of treatment. Whole-cell lysates (50 mg)were analyzed by immunoblotting for the indicated proteins; GAPDH was a loading control. DX-LCLs did not affect T-cell differentiation. C, differentiationstatus of CD3þCD4þ and CD3þCD8þ was assessed by analysis of CCR7 and CD45RA expression in early-stage cultures (day 10), and after the last stimulation(day 35). D, percentages of terminally differentiated (Temra, CCR7�CD45RAþ), effector memory (EM, CCR7�CD45RA�), central memory (CM, CCR7þ

CD45RA�), and na€�ve (CCR7þCD45RAþ) are shown. Left histogram shows CD8þ/CD4þ ratio within CD3þ lymphocytes at day 35. Center and right histogramsshow CD4þ and CD8þ T-cell phenotypes, respectively. Mean of three independent experiments.

Improved Adoptive Cell Therapy for Nasopharyngeal Carcinoma

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 433

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 4: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

EBV-specific CTL differentiation is unaltered by DX-LCLsEBV-specific CTL lines were generated by priming healthy

donor-derived peripheral blood mononuclear cells (PBMC) withautologous LCLs either untreated or treated with TPA þ NaB,doxorubicin, or cisplatin. CD8þ/CD4þ ratio and T-cell differen-tiation profile weremonitored bymultiparametric flow cytometryanalysis at days 10 and 35 (Fig. 1C). The percentage of CD8þ Tcells ranged from 49% to 82% between the first and the laststimulation. Comparable increases in CD8þ T cells were observedat day 35, (67%–77%,�7%),whereasCD4þ cells ranged between6% and 11% (�5%; Fig. 1D). Combined analysis of CCR7 andCD45RA demonstrated that 80% to 90% of CD8þ and 69% to77% of CD4þ T cells displayed an effector memory phenotype(EM, CCR7�CD45RA�; Fig. 1D). Analysis of additional EMmarkers (CD28 and CD62L) demonstrated a high percentage ofCD28þCD8þT cells in all cultures, indicating thatCD8þT cells arenot exhausted. About 50% to 60%ofCD8þ T cells also express theCD62L L-selectin. Analysis of CTL cultures obtained from fourdifferent donors did not reveal any significant difference in thedistribution of CD28 and CD62L in the CCR7�CD45RA�CD8þ

EM population (Supplementary Fig. S1B). Similar findings wereobserved for CD4þ T cells (not shown). Therefore, no significantdifferenceswere observedwith regard to the differentiation profileof the four CTL cultures.

CTLs generated with doxorubicin-treated LCLs kill T2-A2 cellswith exogenous or endogenous BARF1-peptides

EBV-specific CTL cultures were tested for their ability to recog-nize and kill T2-A2 target cells loaded with known EBV-derivedpeptides in a HLA-A�0201–restricted fashion (Fig. 2A and B). Inparticular, T2-A2 cells were loaded with two BARF1-derived pep-tides, p23 and p49 (17), or with the LMP-1–derived YLQ peptide(11). CTLs generated with doxorubicin-treated LCLs (DX-LCLs)were able to specifically kill both BARF1- and LMP-1–loaded T2-A2 inducing a higher percentage of specific lysis as compared withCTLs obtained with TPA þ NaB- or cisplatin-treated LCLs (CSP-LCLs; Fig. 2B). CTL responses against EBNA-1 (FMV) and LMP-2(CLG) EBV peptide epitopes were also investigated. While LMP-2targets were efficiently recognized by bothDX- and cisplatin-CTLs(CSP-CTL; P < 0.05), but not by CTRL- or TPA þ NaB-CTLs,

Figure 2.A, DX-CTLs specifically kill T2-A2 cells loaded with BARF1-derived peptides. Induction of BARF1 (p23 and p49) and LMP1 (YLQ) peptide-specific T-cell responsesfrom three representative out of five HLA-A�0201þ donors [20:1 Effector:target (E:T) ratio]. Specific lysis was calculated by subtracting the lysis of empty T2-A2cells. B, box plot representing cytotoxicity assay performed on T2-A2 cells loaded with BARF1 or LMP-1 peptides. � , P � 0.05 versus the other CTL lines inexperiments carried out on 5 independent HLA-A�0201þ donors. C, DX-CTLs show high specific killing against BARF1-endogenously expressing tumor cells.Cytotoxic activity of CTLs derived from two representative out of five HLA-A�0201þ donors, against c666.1-Wt (negative control) and c666.1-A2, Granta-519,and K562 (specificity control) cells (20:1 E:T ratio). HLA-A�0201 restriction was confirmed with the antibody to HLA-A�0201 cr11.351. D, box plot of cytotoxicityassay results from CTL cultures of five independent HLA-A�0201þ donors tested against BARF1-endogenously expressing tumor cell lines (� , P � 0.05 versusother targets; �� , P � 0.05 versus lysis of other CTLs against c666.1-A2). E, efficiency of CTLs at different E:T ratio.

Fa�e et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology Research434

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 5: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

FMV-loaded cells were lysed at comparable levels by all differentCTL cultures investigated (Supplementary Fig. S1C). To assess theability of DX-CTLs to specifically recognize and kill NPC cellsspontaneously expressing EBV antigens, the HLA-A–negative NPCc666.1 cell line was engineered to stably express HLA-A�0201(c666.1-A2; Supplementary Fig. S2A). As shown in Fig. 2C andD, DX-CTLs were the more efficient and specific cultures in killingNPC cells expressing BARF1 endogenously, whereas no killing wasobserved against the parental, HLA-A–negative c666.1-Wt cells.Conversely, control, TPAþNaB– and CSP-CTLs induced only lowlevelsof specific lysis against c666.1-A2(Fig.2D).The limitedextentof K652 cell killing further ensured the specificity of lysis (Fig. 2E).

EBV-specific cytotoxic activitywas also assessedby enumeratingthe formation of LCL–T cell doublets by multispectral imagingflow cytometry. After incubation of effector T cells with peptide-pulsed autologous LCLs, we observed that DX-CTLs and, partic-ularly, BARF1 peptide–loaded LCLs generated the highest fre-quency of effector–target doublets (Fig. 3A, �, P � 0.05 DX-CTLsvs. all other CTL cultures). Stimulation with the LMP-1 YLQpeptide elicited comparable numbers of doublets in all cultures

except for CTLs generated with untreated LCLs, which showed alower number of doublets (Fig. 3A).

In the same experimental conditions, the intracellular contentof granzyme-b granules was evaluated in all CTL cultures by spot-counting analysis. As shown in Fig. 3B, if compared with CTRL-,TPAþNaB- and CSP-CTLs, a significantly higher absolute numberof granzyme-b granules was detected in DX-CTLs (P � 0.05)stimulated with empty LCLs or with LCLs pulsed with viralpeptides, indicating that DX-LCLs are generally more efficientlyarmed (see the scale of different graphs in Fig. 3B). Notably,DX-CTLs stimulated with BARF1 peptide–pulsed LCLs showedan even higher number of granzyme-b granules as comparedwith those stimulated with LCLs loaded with LMP-1 peptides(P � 0.05; Fig. 3B).

Doxorubicin enhances LCL immunogenicityTo characterize themechanisms underlying the enhanced func-

tional properties ofDX-LCLs as APCs, we investigated the possibleability of doxorubicin to upregulate HLA class I expression.Considering that g-irradiation is currently used to inactivate LCLs

Figure 3.Enumeration of LCL–T cell doublets and granzyme-b granules. Peptide-loaded autologous LCLs were cocultured with DX-CTLs and labeled with antibodies toCD19, CD8, and granzyme-b to enumerate LCL-CTL doublets and granzyme-b–positive spots. A, quantitation of LCL–T cell doublets. The y-axis displays thenormalized frequency of LCL-CTL doublets containing at least one T cell. Mean of four independent experiments performed with four different donor-derivedCTL cultures. � , P � 0.05 DX-CTLs versus all other CTL cultures. Right, representative images (CD19-PE and CD8-PC7), �40 magnification. B, enumeration ofgranzyme-b spots within CD8þ T cells. Left, experiments were performed using DX-CTLs as effector cells and enumerating both the cells and granzyme-bspot formation within these cells. Right, representative images of granzyme-b–FITCþ, CD19-PEþ, and CD8-PC7þ cells. Bottom, granzyme-b granulecontent in Ctrl-, TPA þ NaB-, and cisplatin (CSP)-CTLs. Mean of four independent experiments.

Improved Adoptive Cell Therapy for Nasopharyngeal Carcinoma

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 435

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 6: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

before CTL stimulation and that this procedure was shown toincrease HLA expression (22, 23), we investigated the possiblesynergism between doxorubicin and g-irradiation. HLA-A�0201mRNA expression was therefore monitored by qRT-PCR inLCLs before and 24 hours after g-irradiation (Fig. 4A). DX-LCLsrevealed a marked increase in HLA-A�0201 expression (mean ¼2.3 fold), an effect that persisted also after 24 hours (Fig. 4A). Nosignificant changes were detected in TPA þ NaB– and CSP-LCLscompared with control LCLs (mean ¼ 1.3- and 1.0-fold expres-sion, respectively) at both time points. HLA-A�0201 upregulationin DX-LCLs was also confirmed by flow cytometry analysis at 24hours after g-irradiation (Fig. 4B).

Recent evidence indicates that doxorubicin is able to induceimmunogenic cell death (ICD) both in vitro (24) and in vivo (25,26), an effect mainly mediated by damage-associated molecularpatterns (DAMP), including cell surface–exposed calreticulin(CRT), secreted ATP, and released high mobility group box 1protein (HMGB1; ref. 27). We therefore explored the possibilitythat doxorubicin treatment may induce the expression of mole-cules potentially able to enhance LCL immunogenicity, even atdoses inducing only a limited cell death. Analysis of critical ICDmarkers demonstrated that doxorubicin induces an early (28)CRT exposure (3 hours), with about 50%of cells displaying a CRTmembrane localization. Conversely, only slight increases in CRT

exposure were observed in TPA þ NaB– and CSP-LCLs as com-pared with control LCLs (Fig. 4B). Release of HMGB1 in LCLculturemediumwasmeasured by ELISA at the end of treatment asa late ICD marker (ref. 29; Fig. 4C). Notably, the extent ofHMGB1 release by DX-LCLs was about 3-fold higher than thatof control LCLs, whereas a moderate decrease was observed inboth TPA þ NaB– and CSP-LCLs (about 0.8-fold change).

To verify whether the observed ICD effects persisted or wereenhanced upon g-irradiation, expression of the HSP70 andHSP90 proteins was assessed by immunoblotting immediatelyafter g-irradiation but before coculture with CTLs (t0) and after 24hours (t24; ref. 30; Fig. 4D). Notably, in DX-LCLs, HSP proteinlevels are unchanged after g-irradiation and show a markedincrease at t24. On the contrary, expression levels of both HSP70and HSP90 proteins decrease in TPA þ NaB– and CSP-LCLs at t0and t24, respectively (Fig. 4D). To verify whether the enhancedHMGB1 release showed by DX-LCLs could trigger receptor–dependent responses in an autocrine/paracrine fashion, we eval-uated the expression of its functional receptor, TLR4, and of itsdownstream adaptor molecule Myeloid Differentiation primaryresponse 88 (MyD88; ref. 26). Flow cytometry analysis showedthat doxorubicin and cisplatin markedly increased the number ofTLR4-expressing cells (63.3% and 54.4%, respectively) comparedwith control LCLs (38.1% of positive cells), whereas TPA þ NaB

Figure 4.A, doxorubicin (DX) upregulates HLA-A�0201 expression in LCLs. Relative quantitation by qRT-PCR of HLA-A�0201 mRNA expression in LCLs before (NI, notirradiated; t ¼ 0) and 24 hours after g-irradiation (t24). Data are normalized relative to Ctrl-LCLs. B, HLA-A�02, CRT, and TLR4 cell-surface expression isincreased in DX-LCLs. Expression of HLA-A�02 (top) and TLR4 (bottom) was analyzed by flow cytometry at t24, whereas CRT expression (center) wasinvestigated after 3 hours. Isotype control (dashed line), Ctrl-LCLs (gray area), and treated-LCLs (black line) are shown. C, doxorubicin increases HMGB1 release.Histograms represent the normalized fold increase of HMGB1 released by treated versus untreated LCLs. Experiments were performed in triplicate and doneat least three times on different donor-derived LCLs. D, doxorubicin upregulates HSP-70, HSP-90, and MyD88. Analysis was performed with PARP cleavageimmediately after g-irradiation (t0), and 24 hours after g-irradiation (t24). GAPDH shows equal protein loading. E and F, doxorubicin activates NF-kB anddelays apoptosis onset. Expression of p50, p65, phospho-p65, MyD88, and cleaved caspase-3 was evaluated at the end of doxorubicin treatment; cleaved PARPwas assessed at the end of doxorubicin treatment and at 48 hours.

Fa�e et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology Research436

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 7: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

only slightly affected TLR4 expression (Fig. 4B). In addition,immunoblotting analysis revealed that doxorubicin treatmentupregulates MyD88 expression in LCLs at t0, an effect muchmoreevident at t24 (Fig. 4D), whereas a significant decrease in MyD88protein levels was observed in TPAþNaB- and CSP-LCLs at t0,which became almost undetectable at t24 (Fig. 4D). Moreover,PARP cleavage analysis demonstrated that the late phases of theapoptotic cascade in DX-LCLs occurred only 24 hours afterg-irradiation, whereas a cleaved PARP is already detectable att0 in TPA þ NaB- and CSP-LCLs, which is almost completelyfragmented at t24. To assess the possible role of the TLR4/MyD88pathway in the delayed onset of apoptosis observed for DX-LCLs,we investigated caspase-3 and PARP cleavage, MyD88 expression,and NF-kB activation in DX- LCLs in the presence or absence of aTLR4-neutralizing antibody. Interestingly, the presence of theanti-TLR4 antibody was able to counteract not only the doxoru-bicin-dependent MyD88 upregulation, but also the activation ofsome downstream transcription factors, such as NF-kB, as shownby analysis of the expression of the NF-kB p50 and phospho-p65subunits (Fig. 4E). Furthermore, a higher expression of cleavedcaspase-3 together with slightly decreased levels of the full-lengthPARP form was observed in cells exposed to doxorubicin for 24hours as comparedwith doxorubicin-only LCLs, consistentlywitha delayed onset of the apoptotic cascade in the presence of afunctional TLR4-dependent signaling (Fig. 4E). It is noteworthythat a significant increase of PARP cleavage was observed only48 hours after doxorubicin exposure, an effect that was markedlymore evident in the presence of an anti-TLR4 antibody, support-ing a role for TLR4-dependent signaling in slightly sustainingLCL survival after doxorubicin treatment (Fig. 4E and F).

Lytic activity of DX-CTLs generated from NPC patientsTo validate our results in the NPC setting, we verified whether

doxorubicin was also able to enhance the immunogenicityof LCLs generated from NPC patients. As for healthy donor-derived LCLs, doxorubicin treatment significantly upregulatedBARF1 and LMP-1 mRNA levels (Supplementary Fig. S2B), and

enhanced HLA-A�0201, TLR4 expression, and CRT exposure alsoin LCLs derived from four different NPC patients (Supplemen-tary Fig. S2C). Furthermore, a strong upregulation of the ICDmarkers HSP70, HSP90, and MyD88 was observed in DX-LCLsfrom NPC patients at t0 (Supplementary Fig. S2D). In addition,HMGB1 release was also significantly increased by doxorubicinin patient-derived LCLs as compared with untreated LCLs (Sup-plementary Fig. S2E). To assess the ability of DX-LCLs toefficiently prime and induce EBV-specific effector cells, wegenerated CTLs from four different NPC patients using autol-ogous untreated and DX-LCLs as stimulators. Cytotoxicityassays were performed using T2-A2 cells loaded with BARF1-or LMP-1–derived peptides. As shown in Fig. 5, DX-CTLs dis-played higher specificity and cytotoxic activity to BARF1 pep-tides compared with CTLs induced by untreated LCLs (20%–

40% and 10%–20% specific lysis, respectively). Notably, DX-CTLs from NPC patients also showed an enhanced killing abilityagainst LMP-1–loaded targets (Fig. 5A), further supporting theenhanced immunogenic features of DX-LCLs. More important-ly, DX-CTLs were able to recognize and specifically kill tumorcells expressing BARF1 endogenously, in particular the c666.1-A2 NPC cells and Granta 519 lymphoma cells (50%–60% and20%–30% specific lysis, respectively; Fig. 5B).

DiscussionIn the present study, we report the development of a new and

broadly applicable protocol allowing an efficient generation andexpansionofEBV-specificCTLs enriched in specificities for BARF1,to be used for adoptive immunotherapy of NPC. Exploitation ofthe immunogenicity of this viral protein constitutes a relevantimprovement of current protocols, which were so far designed totarget mainly LMP-1 and LMP-2 proteins, but not BARF1. Indeed,T-cell cultures generated with conventional protocols are virtuallydevoid of BARF1-specific effectors. Considering that BARF1 isexpressed by the majority of NPCs from both endemic and non-endemic areas (13–16), the availability of a protocol able to

Figure 5.DX-CTLs elicit strong EBV-specific responses to both BARF1-and LMP-1–derived peptides, and to BARF1 endogenously expressing NPC cells. A, peptide-specificresponses of CTLs generated by DX-LCLs from 2 HLA-A�0201þNPC patients. Cytotoxicity assays were performed against T2-A2 cells loaded with theindicated peptides (20:1 effector:target ratio); specific lysis was obtained by subtracting empty T2-A2 lysis. B, cytotoxic activity of patient-derived CTLs against thec666.1-Wt, c666.1-A2, Granta-519, and K562 cells. HLA-A�0201 restriction was confirmed with the cr11.351 antibody (not shown). Granta-519 cells may be morevariably lysed andgenerally to a lesser extent than c666.1-A2, consistentlywith the nonconstitutive andmarkedly reduced (about 3–4-fold, not shown) expression ofBARF1 in these cells.

Improved Adoptive Cell Therapy for Nasopharyngeal Carcinoma

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 437

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 8: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

generate EBV-specific CTLs efficiently targeting BARF1 may alsoenhance the still unsatisfactory rates of clinical responses obtain-able by adoptive immunotherapy in this setting (9, 10).

Our protocol exploits the properties of doxorubicin, a com-monly used antineoplastic drug, to promote an abortive EBVreactivation and induce ICD in treated LCLs (31, 32). In fact, wedemonstrate that LCL treatmentwith subcytotoxic concentrationsof doxorubicin is able to upregulate BARF1 expression withoutinducing massive EBV lytic replication, while preserving theexpression of other relevant EBV latency targets, particularlyLMP-1. Consistently, DX-LCLs efficiently generate and expandT-cell effectors able to specifically recognize and kill both autol-ogous targets presenting BARF1-derived epitopes and NPC cellsendogenously expressing BARF1 in an HLA class I–restrictedfashion. Notably, DX-CTLs not only retain the ability to recognizesubdominant LMP-1 epitopes, but also show an increased lyticefficiency against LMP-1–expressing targets as compared witheffectors generated by conventional protocols.

Interesting insights were obtained by counting granzyme-bgranules in EBV-specific T cells, which demonstrated that DX-CTLs include a substantial fraction of effectors readily activatedand "armed" upon stimulation with BARF1 epitope peptides.Intriguingly, also TPA þ NaB– or CSP-CTLs displayed a relativelyhigher number of granzyme-b granule-producing T cells whenthese effectors were cultured in the presence of BARF1-loadedLCLs, consistent with the relative immunodominance of BARF1with respect to LMP-1. Moreover, in the presence of BARF1-presenting targets, DX-CTLs formed the highest number of doub-lets with target LCLs. These findings together support the conclu-sion that DX-CTLs are highly activated effectors with a broaderand more efficient killing potential as compared with T-cellcultures generated with untreated, TPA þ NaB– or CSP-LCLs.

The increased ability of DX-LCLs to generate EBV-specific CTLswith higher efficiency could be at least in part related to theprolonged survival of LCLs as compared with those treated with

other lytic cycle inducers. Notably, persistence of live DX-LCLs incocultures with autologous T lymphocytes is comparable withthat of untreated LCLs, suggesting that treatment with sub-cyto-toxic doses of doxorubicin preserves the LCL integrity necessaryfor an optimal interaction with EBV-specific T-cell precursors.Nevertheless, our results also indicate that other mechanisms areprobably involved in the increased immunogenicity of DX-LCLs.In this respect, our finding that only DX-LCLs show increasedHLA-A�0201 mRNA and protein levels is of particular relevanceconsidering the central role of these molecules in antigen presen-tation. Intriguingly, nanomolar concentrations of doxorubicinwere shown to induce a complex protein ubiquitination responsewithout inhibiting proteasome activities (33), suggesting that theresulting intracellular accumulation of damaged proteins mayinduce quantitative and/or qualitative changes in the immuno-genic epitope repertoire. Our results are therefore consistent withthe possibility that DX-LCLs have enhanced immunogenic prop-erties also because of their ability to induce a more efficient andprolonged presentation of tumor-associated antigen epitopes.Considering that doxorubicin is able to induce ICD (34–36),we explored the hypothesis that doxorubicin could induce func-tional changes presumably responsible for the enhanced immu-nogenic features of LCLs even at doses unable to induce earlyapoptotic effects. Consistent with the ability of doxorubicinto modulate the expression/release of DAMPs both in vitro andin vivo (26), we demonstrate that only doxorubicin was ableto elicit an early CRT exposure at LCL surface, one of the func-tional hallmarks of ICD (35, 37). In the setting of ICD, CRTserves as an "eat me" signal and promotes the immunogenicityof dying tumor cells. In line with the notion that CRT exposureis an early event in ICD, doxorubicin-enhanced CRT exposurewas observed at a time point when no evidence of overt apop-tosis was present. Extracellular HMGB1 is another critical medi-ator of ICD, being released by cells in response to variousstimuli, including proapoptotic drugs (25, 35, 38), and exerting

Figure 6.Proposed mechanisms underlying theenhanced immunogenicity of DX-induced LCLs. Doxorubicin (DX) actsat different levels to improve thefunction of LCLs as APCs for thegeneration of EBV-specific CTLs.Doxorubicin upregulates HLA-A�02surface expression and induces ICD, asshown by enhanced CRT surfaceexposure and HMGB1 release.Doxorubicin also upregulates theexpression of TLR4, thus activating anautocrine/paracrine HMGB1-TLR4loop that enhances LCL survivalthrough NF-kB activation. Thesesurvival signals counteract for acertain period of time theproapoptotic signals induced by thedrug, resulting in a delayed apoptosisof DX-LCLs, which may thus haveprolonged interactions with EBV-specific T-cell precursors. Globally,these effects result in enhancedstimulation and activation of T cells,which also show the ability tospecifically recognize and kill BARF1-expressing tumor cells.

Fa�e et al.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology Research438

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 9: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

direct effects on CD4þ T cells promoting Th1 polarization, T-cellexpansion, and survival. Nonetheless, HMGB1 may also actindirectly by inducing functional changes on APCs. In particular,it has been shown that HMGB1 released by damaged or apoptoticcells may bind to TLR4 on dendritic cells, thereby mediatingdownstream signaling through MyD88 that results in improvedtumor antigen presentation and enhanced CTL activity (38). Ourresults indicate that, besides releasing higher amounts ofHMGB1,DX-LCLs also show increased surface expression of the cognateTLR4 receptor and upregulation of the downstream signalingeffector MyD88, suggesting the occurrence of an autocrine/para-crine loop in the in vitro LCL culture. This local stimulation leads toenhanced NF-kB activation, as shown by upregulation of the p50subunit and enhanced levels of the p65 phosphorylated form.Notably, experiments carried out with a TLR4-neutralizing anti-body demonstrated that both the NF-kB activation and thedelayed onset of apoptosis observed in DX-LCLs, but not in TPAþNaB– or CSP-LCLs are dependent on TLR4 triggering. On thesegrounds, our results support the possibility that the enhancedHMGB1 release induced by doxorubicin may result in the acti-vation of an autocrine/paracrine loop in LCLs able to promote aprolonged survival and an enhanced immunogenicity of theseAPCs (Fig. 6).

We have also investigated the effects of doxorubicin on theexpression of HSP70 and HSP90, key chaperone molecules play-ing important roles in signaling, protein function, trafficking, andturnover (39), andbeing also functionally involved inprotein andepitope folding. Our findings indicate that only doxorubicin, butnot TPA þ NaB– or cisplatin, induces a marked upregulation ofthese proteins, even in the absence of overt apoptosis. Thismay bean additional mechanism underlying the enhanced immunoge-nicity of DX-LCLs and their antigens.

One of the major advantages of our innovative protocolresides in its easy upgradability to GMP standards, consideringthat no molecular engineering of cells is required, and thatdoxorubicin is a drug already and broadly used in the clinics. Itis noteworthy that our protocol does not alter the differenti-ation phenotype of EBV-specific CTLs that are generated andshowed a high efficiency and reproducibility also when PBMCsfrom NPC patients were used. These features are relevant to

ensure the feasibility and the broad applicability of our pro-tocol. We are currently investigating whether the broader anti-gen specificity of DX-CTLs is not restricted to viral proteins butalso involves cellular antigens whose targeting may furtherenhance the therapeutic potential of these effectors.

In conclusion, our results provide the rationale to investigatethe safety and efficacy of DX-LCL–induced EBV-specific CTLs in aphase I/II clinical study of adoptive immunotherapy for NPCpatients. Our final goal is to assess whether our protocol is ableto enhance the rate of clinical responses to adoptive immuno-therapy in NPC patients, particularly for those with relapsed orrefractory disease.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: D.A. Fa�e, D. Martorelli, R. DolcettiDevelopment of methodology: D.A. Fa�e, R. DolcettiAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): D.A. Fa�e, K. Mastorci, E. Muraro, J. Dal Col,G. Franchin, L. Barzan, E. Comaro, E. VaccherAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): D.A. Fa�e, D. Martorelli, K. Mastorci, E. Muraro,J. Dal Col, L. Barzan, R. DolcettiWriting, review, and/or revision of the manuscript: D.A. Fa�e, D. Martorelli,A. Rosato, R. DolcettiAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): D. Martorelli, E. Comaro, A. RosatoStudy supervision: R. Dolcetti

Grant SupportThis study was supported by a grant from the Associazione Italiana per la

Ricerca sul Cancro (contract 14287 toR.Dolcetti). E.Murarowas supported by aFondazione Umberto Veronesi Fellowship.

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 April 17, 2015; revised December 22, 2015; accepted February 7,2016; published OnlineFirst March 23, 2016.

References1. Yu MC, Yuan JM. Epidemiology of nasopharyngeal carcinoma. Semin

Cancer Biol 2002;12:421–9.2. YuX,Wang Z,Mertz JE. ZEB1 regulates the latent-lytic switch in infection by

Epstein-Barr virus. PLoS Pathog 2007;3:e194.3. Chan JKC, Bray F, McCarron P, Foo W, Lee AWM, Yip T, et al. WHO

classification of tumors: pathology and genetics of head and neck tumours.Nasopharyngeal carcinoma . Cedex, France: International Agency forResearch on Cancer (IARC); 2005. p.85–97.

4. Spano JP, Busson P, Atlan D, Bourhis J, Pignon JP, Esteban C, et al.Nasopharyngeal carcinomas: an update. Eur J Cancer 2003;39:2121–35.

5. LoKW, ToKF,HuangDP. Focus onnasopharyngeal carcinoma. CancerCell2004;5:423–8.

6. Huang DP LK. Aetiological factors and pathogenesis. Nasopharyngealcarcinoma . 2nd ed; Chinese University Press; 1999. p.31–60.

7. Merlo A, Turrini R, Dolcetti R, Zanovello P, Amadori A, Rosato A. Adoptivecell therapy against EBV-related malignancies: a survey of clinical results.Expert Opin Biol Ther 2008;8:1265–94.

8. Chia WK, Teo M, Wang WW, Lee B, Ang SF, Tai WM, et al. Adoptive T-cell transfer and chemotherapy in the first-line treatment of metastaticand/or locally recurrent nasopharyngeal carcinoma. Mol Ther 2014;22:132–9.

9. Comoli P, Pedrazzoli P, Maccario R, Basso S, Carminati O, Labirio M, et al.Cell therapy of stage IV nasopharyngeal carcinoma with autologousEpstein-Barr virus-targeted cytotoxic T lymphocytes. J Clin Oncol 2005;23:8942–9.

10. Straathof KC, Bollard CM, Popat U, Huls MH, Lopez T, Morriss MC, et al.Treatment of nasopharyngeal carcinoma with Epstein-Barr virus–specific Tlymphocytes. Blood 2005;105:1898–904.

11. Khanna R, Burrows SR,Nicholls J, Poulsen LM. Identification of cytotoxic Tcell epitopes within Epstein-Barr virus (EBV) oncogene latent membraneprotein 1 (LMP1): evidence for HLA A2 supertype-restricted immunerecognition of EBV-infected cells by LMP1-specific cytotoxic T lympho-cytes. Eur J Immunol 1998;28:451–8.

12. Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, et al.Sustained complete responses in patients with lymphoma receiving autol-ogous cytotoxic T lymphocytes targeting Epstein-Barr virus latent mem-brane proteins. J Clin Oncol 2014;32:798–808.

13. Decaussin G, Sbih-Lammali F, de Turenne-Tessier M, Bouguermouh A,Ooka T. Expression of BARF1 gene encoded by Epstein-Barr virus innasopharyngeal carcinoma biopsies. Cancer Res 2000;60:5584–8.

14. HayesDP, Brink AA, VervoortMB,Middeldorp JM,Meijer CJ, van denBruleAJ. Expression of Epstein-Barr virus (EBV) transcripts encoding

Improved Adoptive Cell Therapy for Nasopharyngeal Carcinoma

www.aacrjournals.org Cancer Immunol Res; 4(5) May 2016 439

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 10: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

homologues to important human proteins in diverse EBV associateddiseases. Mol Pathol 1999;52:97–103.

15. Brink AA, Vervoort MB, Middeldorp JM, Meijer CJ, van den Brule AJ.Nucleic acid sequence-based amplification, a new method for analysis ofspliced and unspliced Epstein-Barr virus latent transcripts, and its com-parison with reverse transcriptase PCR. J Clin Microbiol 1998;36:3164–9.

16. Stevens SJ, Verkuijlen SA,HariwiyantoB,Harijadi, ParamitaDK, Fachiroh J,et al. Noninvasive diagnosis of nasopharyngeal carcinoma: nasopharyn-geal brushings reveal high Epstein-Barr virus DNA load and carcinoma-specific viral BARF1 mRNA. Int J Cancer 2006;119:608–14.

17. Martorelli D, Houali K, Caggiari L, Vaccher E, Barzan L, Franchin G, et al.Spontaneous T cell responses to Epstein-Barr virus-encoded BARF1 proteinand derived peptides in patients with nasopharyngeal carcinoma: bases forimproved immunotherapy. Int J Cancer 2008;123:1100–7.

18. Zhang CX, Decaussin G, Daillie J, Ooka T. Altered expression of twoEpstein-Barr virus early genes localized in BamHI-A in nonproducer Rajicells. J Virol 1988;62:1862–9.

19. Lima RT, Seca H, Bras S, Nascimento MS, Vasconcelos MH. Treatment ofAkata EBV-positive cells with doxorubicin causes more EBV reactivationthan treatment with etoposide. Chemotherapy 2011;57:195–203.

20. Hsu CH, Hergenhahn M, Chuang SE, Yeh PY, Wu TC, Gao M, et al.Induction of Epstein–Barr virus (EBV) reactivation in Raji cells by doxo-rubicin and cisplatin. Anticancer Res 2002;22:4065–71.

21. Countryman JK,Gradoville L,MillerG.Histone hyperacetylationoccurs onpromoters of lytic cycle regulatory genes in Epstein-Barr virus-infected celllines which are refractory to disruption of latency by histone deacetylaseinhibitors. J Virol 2008;82:4706–19.

22. Chiriva-InternatiM,Grizzi F, Pinkston J,MorrowKJ,D'CunhaN, Frezza EE,et al. Gamma-radiation upregulates MHC class I/II and ICAM-I moleculesin multiple myeloma cell lines and primary tumors. In Vitro Cell Dev BiolAnim 2006;42:89–95.

23. Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, WansleyEK, et al. Radiationmodulates the peptide repertoire, enhancesMHCclass Iexpression, and induces successful antitumor immunotherapy. J Exp Med2006;203:1259–71.

24. ApetohL,MignotG, Panaretakis T, KroemerG, Zitvogel L. Immunogenicityof anthracyclines: moving towards more personalized medicine. TrendsMol Med 2008;14:141–51.

25. Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N,et al. Caspase-dependent immunogenicity of doxorubicin-induced tumorcell death. J Exp Med 2005;202:1691–701.

26. Krysko DV, Kaczmarek A, Krysko O, Heyndrickx L, Woznicki J, Bogaert P,et al. TLR-2 and TLR-9 are sensors of apoptosis in a mouse model ofdoxorubicin-induced acute inflammation. Cell Death Differ 2011;18:1316–25.

27. Maccubbin DL, Cohen SA, Ehrke MJ. Indomethacin modulation of adria-mycin-induced effects on multiple cytolytic effector functions. CancerImmunol Immunother 1990;31:373–80.

28. ObeidM, Tesniere A,Ghiringhelli F, FimiaGM,Apetoh L, Perfettini JL, et al.Calreticulin exposure dictates the immunogenicity of cancer cell death.NatMed 2007;13:54–61.

29. Apetoh L,Ghiringhelli F, Tesniere A,ObeidM,Ortiz C, Criollo A, et al. Toll-like receptor 4-dependent contribution of the immune system to antican-cer chemotherapy and radiotherapy. Nat Med 2007;13:1050–9.

30. Garg AD,Martin S, Golab J, Agostinis P. Danger signalling during cancer celldeath: origins, plasticity and regulation. Cell Death Differ 2014;21:26–38.

31. Vacchelli E, Aranda F, Eggermont A, Galon J, Sautes-Fridman C, Cremer I,et al. Trial Watch: Chemotherapy with immunogenic cell death inducers.Oncoimmunology 2014;3:e27878.

32. Mattarollo SR, Loi S, Duret H, Ma Y, Zitvogel L, Smyth MJ. Pivotal role ofinnate and adaptive immunity in anthracycline chemotherapy of estab-lished tumors. Cancer Res 2011;71:4809–20.

33. Mandili G, Khadjavi A, Gallo V, Minero VG, Bessone L, Carta F, et al.Characterization of the protein ubiquitination response induced by Doxo-rubicin. FEBS J 2012;279:2182–91.

34. Ujhazy P, Zaleskis G, Mihich E, Ehrke MJ, Berleth ES. Doxorubicin inducesspecific immune functions and cytokine expression in peritoneal cells.Cancer Immunol Immunother 2003;52:463–72.

35. Zitvogel L, Kepp O, Senovilla L, Menger L, Chaput N, Kroemer G. Immu-nogenic tumor cell death for optimal anticancer therapy: the calreticulinexposure pathway. Clin Cancer Res 2010;16:3100–4.

36. Vacchelli E, Senovilla L, Eggermont A, Fridman WH, Galon J, Zitvogel L,et al. Trial watch: chemotherapy with immunogenic cell death inducers.Oncoimmunology 2013;2:e23510.

37. Garg AD, Dudek AM, Agostinis P. Cancer immunogenicity, danger signals,and DAMPs: what, when, and how?Biofactors 2013;39:355–67.

38. LiG, LiangX, LotzeMT.HMGB1: the central cytokine for all lymphoid cells.Front Immunol 2013;4:68.

39. Pratt WB, Morishima Y, Peng HM, Osawa Y. Proposal for a role of theHsp90/Hsp70-based chaperone machinery in making triage decisionswhen proteins undergo oxidative and toxic damage. Exp Biol Med 2010;235:278–89.

Cancer Immunol Res; 4(5) May 2016 Cancer Immunology Research440

Fa�e et al.

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108

Page 11: Broadening Specificity and Enhancing Cytotoxicity of ... · Research Article Broadening Specificity and Enhancing Cytotoxicity of Adoptive T Cells for Nasopharyngeal Carcinoma Immunotherapy

2016;4:431-440. Published OnlineFirst March 23, 2016.Cancer Immunol Res   Damiana Antonia Faè, Debora Martorelli, Katy Mastorci, et al.   Cells for Nasopharyngeal Carcinoma ImmunotherapyBroadening Specificity and Enhancing Cytotoxicity of Adoptive T

  Updated version

  10.1158/2326-6066.CIR-15-0108doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerimmunolres.aacrjournals.org/content/suppl/2016/03/23/2326-6066.CIR-15-0108.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerimmunolres.aacrjournals.org/content/4/5/431.full#ref-list-1

This article cites 37 articles, 11 of which you can access for free 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/4/5/431To request permission to re-use all or part of this article, use this link

on September 28, 2020. © 2016 American Association for Cancer Research. cancerimmunolres.aacrjournals.org Downloaded from

Published OnlineFirst March 23, 2016; DOI: 10.1158/2326-6066.CIR-15-0108