vaccination with mrna-electroporated dendritic cells · 2012. 9. 10. · cancer therapy: clinical...

12
Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specic CD4 þ and CD8 þ T Cells Responses in Stage III and IV Melanoma Patients Erik H. J. G. Aarntzen 1,2 , Gerty Schreibelt 1 , Kalijn Bol 1,2 , W. Joost Lesterhuis 2 , Alexandra J. Croockewit 3 , Johannes H. W. de Wilt 4 , Michelle M. van Rossum 5 , Willeke A. M. Blokx 6 , Joannes F. M. Jacobs 8 , Tjitske Duiveman-de Boer 1 , Danita H. Schuurhuis 1 , Roel Mus 7 , Kris Thielemans 9 , I. Jolanda M. de Vries 1,2 , Carl G. Figdor 1 , Cornelis J. A. Punt 10 , and Gosse J. Adema 1 Abstract Purpose: Electroporation of dendritic cells (DC) with mRNA encoding tumor-associated antigens (TAA) has multiple advantages compared to peptide loading. We investigated the immunologic and clinical responses to vaccination with mRNA-electroporated DC in stage III and IV melanoma patients. Experimental design: Twenty-six stage III HLA 02:01 melanoma patients scheduled for radical lymph node dissection (stage III) and 19 melanoma patients with irresectable locoregional or distant metastatic disease (referred to as stage IV) were included. Monocyte-derived DC, electroporated with mRNA encoding gp100 and tyrosinase, were pulsed with keyhole limpet hemocyanin and administered intranodally. TAA- specific T-cell responses were monitored in blood and skin-test infiltrating lymphocyte (SKIL) cultures. Results: Comparable numbers of vaccine-induced CD8 þ and/or CD4 þ TAA-specific T-cell responses were detected in SKIL cultures; 17/26 stage III patients and 11/19 stage IV patients. Strikingly, in this population, TAA-specific CD8 þ T cells that recognize multiple epitopes and produce elevated levels of IFNg upon antigenic challenge in vitro, were significantly more often observed in stage III patients; 15/17 versus 3/11 stage IV patients, P ¼ 0.0033. In stage IV patients, one mixed and one partial response were documented. The presence or absence of IFNg -producing TAA-specific CD8 þ T cells in stage IV patients was associated with marked difference in median overall survival of 24.1 months versus 11.0 months, respectively. Conclusion: Vaccination with mRNA-electroporated DC induces a broad repertoire of IFNg producing TAA-specific CD8 þ and CD4 þ T-cell responses, particularly in stage III melanoma patients. Clin Cancer Res; 18(19); 1–11. Ó2012 AACR. Introduction Dendritic cells (DC) are the most effective antigen-pre- senting cells (APC) of the immune system, highly capable of stimulating naive T cells. Immunotherapy with ex vivo- generated autologous DC pulsed with tumor peptides has provided proof of concept in clinical trials (1). We, and others, have showed that tumor-specific immune responses can be induced in both stage III and IV melanoma patients (1–5). Because objective clinical responses are observed in a minority of patients, further optimization of DC-based immunotherapy is warranted. To date, the majority of clinical studies on DC-based vaccinations have been conducted with MHC class I restrict- ed peptide-pulsed monocyte-derived DC in patients with measurable distant metastatic disease. However, there are at least several theoretical disadvantages against these proto- cols, which might be improved to induce more effective and sustained immunologic responses. First, the exploitation of MHC class I restricted peptide epitopes target CD8 þ CTL only, without involving CD4 þ T helper cells to enhance and sustain antitumor CTL responses. Second, pulsing DC with peptide epitopes implicates the use of a given HLA type, with defined tumor-associated antigens (TAA). Moreover, peptide-loaded DC expose the antigen only for a short period of time (6), because the peptides may readily dis- sociate from the MHC molecules (7). Importantly, peptide Authors' Afliations: 1 Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences; 2 Departments of Medical Oncology, 3 Hematology, 4 Surgery, 5 Dermatology, 6 Pathology, 7 Radiology, 8 Depar- ment of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands; 9 Laboratory of Molecular & Cellular Therapy, Department of Physiology-Immunology, Vrije Universiteit Brussel, Brussel, Belgium; and 10 Department of Medical Oncology, Academic Medical Centre, Amsterdam, The Netherlands Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). E.H.J.G. Aarntzen and G. Schreibelt have contributed equally to this work. Corresponding Author: Gosse J. Adema, Department of Tumor Immu- nology, Nijmegen Centre for Molecular Life Sciences, PO Box 9101, 6500 HB Nijmegen, The Netherlands. Phone: 31-24-3617600; Fax: 31-24- 3540339; Email: [email protected] doi: 10.1158/1078-0432.CCR-11-3368 Ó2012 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org OF1 Cancer Research. on November 24, 2020. © 2012 American Association for clincancerres.aacrjournals.org Downloaded from Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Upload: others

Post on 17-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

Cancer Therapy: Clinical

Vaccination with mRNA-Electroporated Dendritic CellsInduces Robust Tumor Antigen-Specific CD4þ and CD8þ

T Cells Responses in Stage III and IV Melanoma Patients

Erik H. J. G. Aarntzen1,2, Gerty Schreibelt1, Kalijn Bol1,2, W. Joost Lesterhuis2, Alexandra J. Croockewit3,Johannes H. W. de Wilt4, Michelle M. van Rossum5, Willeke A. M. Blokx6, Joannes F. M. Jacobs8,Tjitske Duiveman-de Boer1, Danita H. Schuurhuis1, Roel Mus7, Kris Thielemans9, I. Jolanda M. de Vries1,2,Carl G. Figdor1, Cornelis J. A. Punt10, and Gosse J. Adema1

AbstractPurpose: Electroporation of dendritic cells (DC) withmRNA encoding tumor-associated antigens (TAA)

has multiple advantages compared to peptide loading. We investigated the immunologic and clinical

responses to vaccination with mRNA-electroporated DC in stage III and IV melanoma patients.

Experimental design: Twenty-six stage III HLA�02:01 melanoma patients scheduled for radical lymph

node dissection (stage III) and 19 melanoma patients with irresectable locoregional or distant metastatic

disease (referred to as stage IV) were included. Monocyte-derived DC, electroporated withmRNA encoding

gp100 and tyrosinase, were pulsed with keyhole limpet hemocyanin and administered intranodally. TAA-

specific T-cell responses were monitored in blood and skin-test infiltrating lymphocyte (SKIL) cultures.

Results:Comparablenumbers of vaccine-inducedCD8þ and/orCD4þTAA-specific T-cell responseswere

detected in SKIL cultures; 17/26 stage III patients and 11/19 stage IV patients. Strikingly, in this population,

TAA-specific CD8þ T cells that recognize multiple epitopes and produce elevated levels of IFNg upon

antigenic challenge in vitro, were significantly more often observed in stage III patients; 15/17 versus 3/11

stage IVpatients,P¼0.0033. In stage IVpatients, onemixed andonepartial responsewere documented. The

presence or absence of IFNg-producing TAA-specific CD8þ T cells in stage IV patients was associated with

marked difference in median overall survival of 24.1 months versus 11.0 months, respectively.

Conclusion: Vaccination with mRNA-electroporated DC induces a broad repertoire of IFNg producingTAA-specific CD8þ and CD4þ T-cell responses, particularly in stage III melanoma patients. Clin Cancer Res;

18(19); 1–11. �2012 AACR.

IntroductionDendritic cells (DC) are the most effective antigen-pre-

senting cells (APC)of the immune system, highly capable ofstimulating naive T cells. Immunotherapy with ex vivo-

generated autologous DC pulsed with tumor peptides hasprovided proof of concept in clinical trials (1). We, andothers, have showed that tumor-specific immune responsescan be induced in both stage III and IV melanoma patients(1–5). Because objective clinical responses are observed in aminority of patients, further optimization of DC-basedimmunotherapy is warranted.

To date, the majority of clinical studies on DC-basedvaccinations have been conductedwithMHC class I restrict-ed peptide-pulsed monocyte-derived DC in patients withmeasurable distantmetastatic disease. However, there are atleast several theoretical disadvantages against these proto-cols, whichmight be improved to inducemore effective andsustained immunologic responses. First, the exploitation ofMHC class I restricted peptide epitopes target CD8þ CTLonly, without involving CD4þ T helper cells to enhance andsustain antitumor CTL responses. Second, pulsing DC withpeptide epitopes implicates the use of a given HLA type,with defined tumor-associated antigens (TAA). Moreover,peptide-loaded DC expose the antigen only for a shortperiod of time (6), because the peptides may readily dis-sociate from the MHC molecules (7). Importantly, peptide

Authors' Affiliations: 1Department of Tumor Immunology, NijmegenCentre for Molecular Life Sciences; 2Departments of Medical Oncology,3Hematology, 4Surgery, 5Dermatology, 6Pathology, 7Radiology, 8Depar-ment of Laboratory Medicine, Radboud University Nijmegen MedicalCentre, Nijmegen, The Netherlands; 9Laboratory of Molecular & CellularTherapy,Department of Physiology-Immunology, VrijeUniversiteit Brussel,Brussel, Belgium; and 10Department of Medical Oncology, AcademicMedical Centre, Amsterdam, The Netherlands

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

E.H.J.G. Aarntzen and G. Schreibelt have contributed equally to this work.

Corresponding Author: Gosse J. Adema, Department of Tumor Immu-nology, Nijmegen Centre for Molecular Life Sciences, PO Box 9101, 6500HB Nijmegen, The Netherlands. Phone: 31-24-3617600; Fax: 31-24-3540339; Email: [email protected]

doi: 10.1158/1078-0432.CCR-11-3368

�2012 American Association for Cancer Research.

ClinicalCancer

Research

www.aacrjournals.org OF1

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 2: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

loading does not account for posttranscriptional modifica-tions of peptide epitopes (8, 9).

One strategy to circumventmost of these disadvantages ofpeptide pulsing is electroporation with synthetic mRNAencoding TAA, resulting in endogenous synthesis of thecomplete TAA. It has been shown previously that electro-poration of DC with mRNA is effective and safe (7, 10, 11).DCs retain their phenotype and maturation potentialupon electroporation, as well as their migratory capacities(10, 12). Electroporated DC express TAA antigens, encodedby the electroporated mRNA and induce specific CD8þ T-cell responses in melanoma patients (10). Importantly,because mRNA lacks the potential to integrate into the hostgenome, it obviates safety concerns associated with genetherapy trials.

It is now widely recognized that high tumorload inpatients with end-stage cancer often induces local, or evensystemic, immune suppression by the secretion of suppres-sive cytokines and attraction of regulatory T cells (13–15).This suppressive tumor microenvironment will hamper theeffective antitumor responses. Melanoma patients withlocoregional lymph node metastases are at high risk ofrelapse and currently no standard adjuvant treatment isavailable which results in overall survival benefit (16).Given the minimal burden of tumor, we hypothesized thatvaccination of patients adjuvant to therapeutic radicallymph node dissection might enhance vaccine efficacy.

In this study we investigated in detail the immunologicresponses to intranodal vaccinationwithmonocyte-derivedDC electroporated with mRNA encoding gp100 and tyros-inase in 2 cohorts of melanoma patients; with distantmetastatic or irresectable locoregional disease followingradical regional lymph node dissection.

Patients and MethodsPatient population

Melanoma patients with locoregional resectable disease(further referred to as stage III), before or within 2 monthsafter radical dissection of regional lymph node metastases,and patients with irresectable locoregional or distant met-astatic disease (further referred to as stage IV)were included.Additional inclusion criteria were HLA�02:01 phenotype,melanoma expressing the melanoma-associated antigensgp100 and tyrosinase, and World Health Organizationperformance status 0 or 1. Patients with brain metastases,serious concomitant disease, or a history of a secondmalig-nancy were excluded. The study was approved by ourInstitutional Review Board, and written informed consentwas obtained from all patients. Clinical trial registrationnumber is NCT00243529.

Study protocolPatients received a DC vaccine intranodally, injected into

a clinically tumor-free lymph node under ultrasound guid-ance. The DC vaccine consisted of autologous maturemonocyte-derivedDC electroporatedwithmRNAencodingfor gp100 and tyrosinase protein, and pulsed with keyholelimped hemocyan (KLH) protein. Patients received 3 vac-cinations with a biweekly interval. Ten patients received anextra vaccination 1 or 2 days before the radical lymph nodedissection for additional imaging studies (manuscript inpreparation).One to 2weeks after the last vaccination a skintest was conducted. In absence of disease progression orrecurrence, patients received a maximum of 2 maintenanceseries at 6-month intervals, each consisting of 3 biweeklyintranodal vaccinations (Supplementary Fig. S1). All vacci-nations were administered between May 2006 and May2010. Patients were considered evaluable when they hadcompleted the first vaccination cycle. Vaccine-specificimmune response was the primary endpoint, clinicalresponse was a secondary endpoint in stage IV patients.Progression-free and overall survival were calculated fromthe time from apheresis to recurrence (for stage III patients)or progression (for stage IV patients), or death.

DC preparation and characterizationDC were generated from peripheral blood mononuclear

cells (PBMC) prepared from leukapheresis products asdescribed previously (17). After leukapheresis, part of thePBMC was used for the generation of monocyte-condi-tioned medium (MCM; 18). Plastic-adherent monocytesor monocytes isolated by centrifugal elutriation were cul-tured for 5 to 7 days in X-VIVO 15 medium (BioWhittaker)supplemented with 2% pooled human serum (HS) (Blood-bank Rivierenland), interleukin 4 (IL-4; 500 U/mL), andgranulocyte macrophage colony-stimulating factor (800 U/mL; both fromCellgenix). Immature DCwere pulsed at day3 with KLH (10 mg/mL; Calbiochem). Two days before theharvesting, cells werematuredwith autologousMCM, pros-taglandin E2 (10 mg/mL; Pharmacia & Upjohn) and recom-binant tumor necrosis factor alpha (10 ng/mL; provided by

Translational RelevanceElectroporation of dendritic cells (DC) with mRNA

encoding tumor-associated antigens hasmultiple advan-tages compared to the conventional peptide loading.The presentation of multiple naturally processed epi-topes in both MHC Class I and II should broaden therepertoire of responding lymphocytes.We studied in detail the immunologic response to

vaccination with mRNA-electroporated DC in 2 cohortsof melanoma patients: as palliative treatment of distantor irresectable locoregional metastatic disease and asadjuvant treatment following radical dissection ofregional lymph nodes. A wide spectrum of tumor-spe-cific IFNg producing CD8þ T cells was detected, inparticular in patients vaccinated in the adjuvant setting.Furthermore, vaccine-induced CD4þ T cells were shownto be FoxP3 negative.In conclusion, vaccination with mRNA-electropo-

rated DC successfully enhances antitumor cytotoxic T-cell responses and appears to be a promising adjuvanttreatment of stage III melanoma patients.

Aarntzen et al.

Clin Cancer Res; 18(19) October 1, 2012 Clinical Cancer ResearchOF2

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 3: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

Dr. G. Adolf, Bender Wien; 19). This protocol gave increaseto amature phenotypemeeting the release criteria describedpreviously (20): low expressionofCD14, high expression ofMHC class I, MHC class II, CD83, CD80, CD86, and CCR7,and expression of gp100 and tyrosinase after electropora-tion with mRNA (Supplementary Fig. S2). Harvested DCwere tested by fluorescence-activated cell sorting (FACS)analysis as described below.

Plasmids and in vitro mRNA transcriptionPlasmids have been sent to CureVac GmbH for the

production of documented GMP grade gp100 and tyrosi-nase RNA for ex vivo use in clinical DC vaccination. Thedocumented gp100 and tyrosinase mRNA was producedfrom the plasmids pGEM4Z/hgp100/A64 and pGEM4Z/tyrosinase/A64 (provided by Kris Thielemans, Free Univer-sity Brussels, Belgium) according to GMP guidelines. Cur-eVacmRNA contains a 50 cap and 30 poly A-tail that leads tohigh-RNA stability and increased protein expression intransfected cells. The mRNA is purified by PUREmessengertechnology. This chromatography method efficiently era-dicates traces of DNA and proteins. The mRNA productionprocess is carried out in clean room facilities and is docu-mented by in-process controls. RNA quality was verified byagarose gel electrophoresis, RNA concentration was mea-sured spectrophotometrically, and RNA was stored at�80�C in small aliquots.

Electroporation of DCMature DC were electroporated as described previously

(10). Briefly, DC were washed twice in PBS and once inOptiMEM without phenol red (Invitrogen). Twenty micro-grams of RNA encoding either gp100 or tyrosinase weretransferred to a 4-mm cuvette (Bio-Rad) and 8 � 106 cellswere added in 200 mLOptiMEM and incubated for 30 beforebeing pulsed in a Genepulser Xcell (Bio-Rad) by an expo-nential decay pulse of 300 V, 150 mF, as described before(10). Immediately after electroporation, cells were washedand were transferred to warm (37oC) X-VIVO 15 withoutphenol red (Cambrex Bio Science) supplemented with 5%HS and left for at least 2 hours at 37oC, before furthermanipulation. The first vaccinationwas givenwith freshDC4 hours after electroporation. DC for subsequent vaccina-tions were frozen 2 hours after electroporation, thawed atthe day of vaccination, and incubated for 2 more hours at37�C before injection. Electroporation efficiency was ana-lyzed by intracellular staining and flow cytometric analysisfor each separate TAA, electroporatedDCweremixedbeforevaccination.

Flow cytometric analysisThe following fluorescein isothiocyanate-conjugated

mAbs were used: anti-HLA class I (W6/32), and anti-HLADR/DP (Q5/13); and PE-conjugatedmAbs: anti-CD80 (BDBiosciences), anti-CD14, anti-CD83 (both Beckman Coul-ter), and anti-CD83 (BD Pharmingen). For intracellularstaining of the TAA the following mAb were used: NKI/beteb (IgG2b; purified antibody) against gp100, T311

(IgG2a; Cell Marque Corp.) against tyrosinase. For intra-cellular staining cells were fixed for 40 on ice in 4% (w/v)paraformaldehyde (Merck) in PBS, permeabilized in PBS/2%BSA/0.02% azide/0.5% saponin (Sigma-Aldrich; PBA/saponin), and stained with mAb diluted in PBA/saponin/2%HS, followed by staining with allophycocyanin-labeledgoat-anti-mouse (BD PharMingen). Flow cytometry wasconductedwith FACSCalibur flow cytometer equippedwithCellQuest software (BD Biosciences).

Flow cytometric analysis of T cells was conducted usingdirectly labeled mAbs against CD4, CD8, CD25, CD127,CTLA-4 (BD Pharmingen), and FoxP3 (eBiosciece), allaccording to the manufacturer’s protocol. Tregs weredefined as CD4þFoxP3þCD25highCD127low cells; percent-age of Tregs was defined as the number of CD4þ

FoxP3þCD25highCD127low cells divided by the total num-ber of CD4þ cells �100.

KLH-specific proliferationKLH-specific cellular responses were measured before

and after vaccination by proliferation assay. PBMC wereisolated from heparinized blood by Ficoll–Paque densitycentrifugation. PBMC were stimulated with KLH (4 mg/2 �105 PBMC) in mediumwith 10% human AB serum. After 3days, cells were pulsed with 3H-thymidine for 8 hours, andincorporation was measured with a betacounter. Experi-ments were conducted in triplicate.

KLH-specific antibody productionKLH-specific antibodies were measured in the sera of

patients before and after vaccination. Microtiter plates (96wells) were coated overnight at 4oC with KLH (25 mg/mL inPBS). Different concentrations of patient serum (range1:100 to 1:50,000) were added for 600 at room temperature.After extensive washing, patient antibodies were detectedwithmouse anti-human IgG, IgA, or IgM antibodies labeledwith horseradish peroxidase (Invitrogen). 3,30 5,5-tetra-methyl-benzidine was used as a substrate and plates weremeasured with a microtiter plate reader at 450 nm. Forquantification, an isotype-specific calibration curve for theKLH response was included in each microtiter plate.

Skin-test infiltrating lymphocyte culturesNote that 0.2� 1� 106DCpulsedwith the gp100 and/or

tyrosinase peptides andDC electroporatedwith gp100 and/or tyrosinase mRNA each were injected intradermally in theskin of the back of the patient at 4 or 6 different sites (21).The maximum diameter of induration was measured after48 hours.1 From each site induration was measured andpunch biopsies (6 mm) were obtained. Half of the biopsywas cryopreserved and the other part was manually cutand cultured in RPMI 1640 containing 7% HS and IL-2(100 U/mL). Every 7 days, half of the mediumwas replacedby freshmedium containing HS and IL-2. After 2 to 4 weeks

1A detailed description of the DTH procedure can be found at http://www.labtube.tv/playvideo.aspx?vid¼131825

Vaccination of Melanoma Patients with mRNA-Electroporated DC

www.aacrjournals.org Clin Cancer Res; 18(19) October 1, 2012 OF3

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 4: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

of culturing, skin-test infiltrating lymphocytes (SKIL)were tested. In general, similar results were obtained perpatient, regardless of the method of antigen-presentation(for example, Supplementary Fig. S3).

Tetramer stainingSKILs and freshly isolated PBMC were stained with tet-

rameric-MHC complexes containing the HLA-A2-bindingepitopes gp100:154–168, gp100:280–288, or tyrosi-nase:369–377 (Sanquin) or HLA-DR4-binding epitopesgp100:44–59 and tyrosinase:448–462 (provided by Wil-liam Kwok, Benaroya Research Institute, Seattle, WA) asdescribed previously (19). In addition, PBMCs of patientswith tetramer positive CD4þ T cells were restimulated for8 days with DR4-binding gp100 or tyrosinase peptidesand stained with tetrameric-MHC complexes containingclass II epitopes gp100:44–59 and tyrosinase:448–462.Tetrameric-MHC complexes recognizing HIV were used ascontrols; at least a 2-fold increase of the double-positivepopulation compared with control was regarded to bepositive.

Antigen and tumor recognitionAntigen recognition was determined by the production

of cytokines and cytotoxic activity of SKILs in response toT2 pulsed with the indicated peptides or BLM (amelanomacell line expressing HLA�02:01 but no endogenous expres-sion of gp100 and tyrosinase), transfected with controlantigen G250, with gp100 or tyrosinase, or an allogenicHLA�02:01-positive, gp100-positive, and tyrosinase-posi-tive tumor cell line (MEL624) were measured. Cytokineproduction was measured in supernatants after 16 hours ofcoculture by a cytrometric bead array (Th1/Th2 CytokineCBA 1; BD PharMingen). Positive and specific cytokineproduction was defined as a 2-fold increase compared withstimulation with the cell lines pulsed with an irrelevantpeptide.

Statistical analysisDifferences between the groups were evaluated using the

Fisher exact test or one-way ANOVA. Statistical significancewas defined as P < 0.05. GraphPad Prism5.0was used for allanalyses.

ResultsPatient characteristics

A total of 48HLA�02:01positivemelanomapatients wereenrolled (Supplementary Fig, S1), of which 3 patients (IV-D-13, IV-D-12, IV-D-07) were regarded as nonevaluable,because they did not complete the first cycle because ofrapid progressive disease. Two stage IV patients were onlyevaluable for immunologic response; patient IV-C-05 hadno measurable disease at baseline and patient IV-D-15 hadproven brain metastasis after the second vaccination butcompleted the first cycle. Twenty-six stage III and 19 stage IVpatients were included. Twenty-three stage III patientsreceived 1 cycle of maintenance vaccinations and 20

patients completed the full 3 cycles. Three stage IV patientsreceived 1 cycle of maintenance vaccinations; 1 patientcompleted the full 3 vaccination cycles. No unexpected orsevere adverse events were observed. Patient characteristicsare summarized in Table 1.

Vaccine characteristicsPhenotypic and functional release criteria were defined

for DC vaccines to ensure minimal quality criteria and theusage of mature DC in clinical vaccination protocols (22).The phenotype of the ex vivo-generated DC was determinedby flow cytometry and all produced vaccines met the stan-dard release criteria, with respect to expression ofMHC classI and II, and costimulatory molecules, CD83 and CCR7(Supplementary Fig. S2A). Furthermore, we confirmed theintracellular expression of TAAs gp100 and tyrosinase afterelectroporation by flow cytometry (Supplementary Fig.S2B). Patients received on average 12 � 106 DC per vacci-nation with a maximum of 15 � 106 DC per vaccination.

Immunologic response to KLHFor immunomonitoring purposes all DC have been

loaded with the control antigen KLH. PBMC, isolated aftereach vaccination, showed increased proliferation uponstimulation with KLH after vaccination in almost allpatients in the first cycle (Fig. 1A). One patient first devel-oped a proliferative response to KLH in the second cycle.Anti-KLH antibodies were detected in 9 out of 17 stage IVpatients tested, and 15out of 26 stage III patients tested (Fig.1B). These data show that the vaccine effectively induced denovo immune responses.

TAA-specific responses in bloodTo investigate TAA-specific immune responses, PBMC

were screened with tetrameric-MHC complexes before andafter each cycle of 3 vaccinations at the time point of SKILtest. TAA-specific CD8þ T cells were only found in freshlyisolated PBMC from 3 stage III and 3 stage IV patients aftervaccination (Fig. 2 and Table 2). Because it has beendescribed that melanoma patients can already have a sub-stantial number of TAA-specific T cells circulating in theirblood, we analyzed the presence of TAA-specific T cells inPBMC isolated before vaccination. Three out of these 6patients had no detectable TAA-specific CD8þ T cells circu-lating before vaccination, suggesting that TAA-specificCD8þ T cells were newly induced, or at least enhanced, bythe DC vaccinations in these patients, in concordance withprevious reports (23).

Evidence is emerging that CD4þ T cells need to beantigen-specific to potentiate the CD8þ immune response.Therefore we analyzed the presence of TAA-specific CD4þ Tcells in PBMCs of all 15 DR4þ patients. Four patients werepositive after vaccination (patients IV-C-01, IV-C-02, IV-C-03, IV-C-08; Fig. 3A and Table 2). Tetramer analysis ofPBMCs restimulated in vitro with DR4-binding peptidesconfirmed the presence of TAA-specific CD4þ T cells (Fig.3B). TAA-specific CD4þ T cells were detectable before vac-cination in only 1 patient (IV-C-03), but only after in vitro

Aarntzen et al.

Clin Cancer Res; 18(19) October 1, 2012 Clinical Cancer ResearchOF4

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 5: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

Table 1. Patient characteristics

Patient SexAge(yrs)

Origin ofprimarytumor

Stage onentry

Site ofmetastaticdisease

LDH(U/L) gp100c Tyroc

HLA-DR4status

# of DC pervaccination �106

# ofvaccinations

Previoussystemictreatment

Salvagesystemictreatment

Stage IV Mean Min MaxII-E-01 M 54 Skin M1c LN 542 þþ þ – 8,8 6 13 3 No DTICIV-C-01 F 60 Skin M1b Lung, LN 343 þþþ þþþ þ 10,7 9 12 3 No DTICIV-C-02 F 48 Skin M1c Liver, lung, skin 318 þþþ þþ þ 11,0 8 14 3 No TMZIV-C-05 F 31 Skin M1c Lung, skin, breast 301 þþþ þ þ 14,0 9 15 12 No NoIV-C-06 M 57 Skin M1c Lung, bladder, skin 412 þþ – þ 12,3 11 15 3 No NoIV-C-09 F 48 Skin M1b Lung, skin, LN 402 þþþ – þ 12,0 10 15 3 DTIC NoIV-C-13 F 47 Skin M1c Skin, LN 531 þ þ þ 12,3 9 15 3 DTIC NoIV-D-01 M 75 Skin M1c Liver, lung, bladder 334 þþþ þþþ – 11,5 11 12 3 DTIC NoIV-D-02 M 69 Skin M1c Liver, lung, bone,

adrenal502 þ þþ – 10,0 7 12 3 No No

IV-D-03 F 42 Eye M1c Liver, lung 344 þþþ þ – 7,3 6 9 3 DTIC TMZIV-D-04 M 59 Skin M1c Lung, bladder, LN 381 þþþ þþ – 9,7 8 12 3 No DTIC, TMZ,

AZD6244IV-D-06 F 61 Skin M1a LN 459a þ þ – 11,5 5 15 6 No NoIV-D-09 M 50 Skin M1a LN 431 þþþ þþþ – 15,0 15 15 3 No DTICIV-D-11 M 44 Mucosa N3 LN, maxillary sinus 351 þþþ þþ – 14,2 12 15 12 No NoIV-D-15 M 58 Skin M1c Lung, bone, LN 523 þþþ þþþ – 14,7 14 15 3 No DTICIV-D-17 M 66 Skin M1c Lung, adrenal,

intracardial387 þþþ þþþ – 15,0 15 15 3 No No

IV-D-18 M 57 Skin M1a Skin, LN 369 þþþ þþþ – 13,0 11 15 6 No DTIC,AZD6244

IV-D-20 M 62 Skin M1c Liver, LN, adrenal 390 þþþ þ – 15,0 15 15 3 No DTICIV-D-21 F 30 Skin M1c Liver, lung, LN, skin 421 þþþ þþþ – 9,3 5 15 3 No DTICStage IIIII-E-02 M 38 Skin N3 LN 368 þþþ þþ – 11,3 6 15 12 NoII-E-03 M 45 Skin N1a LN 351 þþþ þþ þ 13,9 10 15 12II-E-04 M 41 Skin N2a LN 392 þþþ þ – 6,8 3 11 12II-E-05 F 49 Skin N2a LN 279 – þþ – 11,2 6 15 12 DTICII-E-06 M 48 Skin N2a LN 356 þþþ þþ – 12,6 10 15 12 NoII-E-07 F 59 Skin N3 LN 422 þ n.a. – 14,1 12 15 12II-E-08 M 56 Skin N3 LN 598a þþ þþ – 12,0 9 15 3 DTICII-E-09 M 26 Skin N3 LN 352 þþþ þþþ – 13,5 10 15 12 UnknownII-E-10 F 34 Skin N1a LN 348 þþþ þþþ – 14,5 13 15 12II-E-11 M 47 Skin N1a LN 299 þþþ þþ – 13,8 10 15 12IV-C-03 M 37 Skin N1a LN 393 þþþ þþþ þ 12,2 8 15 12 DTICIV-C-04 M 45 Skin N3 LN 408 þ þ þ 9,0 8 10 7 NoIV-C-07 M 35 Skin N1a LN 335 þþþ þþþ þ 13,0 9 15 12IV-C-08 M 35 Skin N1a LN 390 þþ þþ þ 14,2 8 15 12IV-C-10 F 36 Skin N1a LN 361 þþþ þþ þ 13,1 9 15 12IV-C-11 M 50 No primary N1b LN 379 þ þ þ 14,0 11 15 12IV-C-12 F 38 No primary N1b LN 360 þþþ þþþ þ 13,7 11 15 3 DTICIV-C-14 F 54 Skin N1b LN 458 þþþ þ þ 12,2 7 15 12IV-D-05 M 48 Skin N3 LN 388 þþ þþ – 7,0 6 8 6 DTICIV-D-08 F 54 Skin N2b LN 416 þþþ þ – 15,0 15 15 6 NoIV-D-10 F 37 Skin N1a LN 342 þþ þþ – 11,6 10 15 12 NoIV-D-14 M 63 Skin Nxb LN 348 þþþ þ – 13,3 10 15 12IV-D-16 M 31 Skin N2b LN 517a þþþ þþþ – 12,1 8 15 12IV-D-19 M 62 Skin N1b LN 280 þþþ þ – 10,8 7 15 12 ITxIV-D-22 M 23 Skin N1b LN 394 þþ – – 13,3 12 15 3 DTIC,

anti-CTLA4IV-D-24 M 65 Skin N2a LN 350 þþ þþ – 14,2 12 15 12 No

Abbreviations: DTIC, dacarbazine; F, female; LDH, lactate dehydrogenase; LN, lymphnode;M,male; nr, number, TMZ, Temozolomide.aLDH normalized after RLND.bPatients stopped because of burden of skin testcgp100and tyrosinaseexpressionon theprimary tumorwas analyzedby immunohistochemistry. Intensity of positive cellswere scoredcentrally and semiquantitatively by a pathologist. Intensity was scored as low (þ), intermediate (þþ), or high (þþþ).

Vaccination of Melanoma Patients with mRNA-Electroporated DC

www.aacrjournals.org Clin Cancer Res; 18(19) October 1, 2012 OF5

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 6: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

restimulation of PBMCs with DR4-binding peptides, sug-gesting that tumor-specific CD4þ T cells were induced, or atleast enhanced, by DC vaccinations in these patients. Weidentified concurrent TAA-specific CD8þ T cells in SKILcultures in 3 of the 4 patients with tumor-specific CD4þ

T cells in their blood (patients IV-C-01, IV-C-02, IV-C-03; Table 2). To exclude that the TAA-specific CD4þ T cellshave a suppressor phenotype, we tested their FoxP3 expres-sion, all were negative (Fig. 3C).

TAA-specific responses in SKIL culturesPreviously we showed that the presence of TAA-specific T

cells in SKIL cultures positively correlates with clinicaloutcome in stage IV melanoma patients (22). Skin testswere carried out after each cycle of vaccinations. Becauseconducting skin tests and taking biopsies puts a greatburden to the patient, and previously we observed in aseries of patients who underwent prevaccination skin testanalysis that none of the patients had detectable levels ofTAA-specific T cells before vaccination, we choose not to

conduct prevaccination skin tests, but rather conduct in-depth analysis on the postvaccination samples. Tetramerpositive CD8þ SKILs were detected in 17 stage III patientsand in 11 stage IV patients (P¼ 0.7574 Fischer exact test). In8 stage III patients and in 2 stage IV patients, CD8þ SKILswere specific for all 3 tested epitopes (Table 2, Fig. 4A). Sixstage III and 3 stage IV patients had TAA-specific CD8þ Tcells against 2 of the 3 epitopes tested, although CD8þ Tcells of the other patients recognized 1 epitope.

Merely the presence of TAA-specific CD8þ T cells is notnecessary sufficient for effective antitumor responses. There-fore, we tested whether the vaccine-induced TAA-specificCD8þ T cells were "functional" in terms of selective IFNgproduction upon coculture with peptide-, or protein-load-ed target cells or tumor cells (Table 2, Fig. 4B). Strikingly,althoughwe detected tetramer-specific CD8þ T cells in bothstage III and IV melanoma patients to similar extend, wefound increased IFNg productiononly in3out of 11 stage IVmelanoma patients. In contrast, in stage III patients with nomeasurable disease, we found IFNg production in 15 out of17 patients with tetramer-positive CD8þ SKIL cultures, (P¼0.0033 Fisher exact test).

Interestingly, SKILs of 3 patients (IV-C-02, IV-C-10, IV-C-14) that did not produce cytokines upon coculture with theHLA�02:01 binding peptides, produced IFNg upon cocul-turewith the respective tumorprotein, indicating that T cellsrecognized different epitopes. SKILs derived from threeadditional patients (II-E-04, II-E-05, IV-C-12) that pro-duced IFNg upon stimulation with only one of theHLA�02:01 binding peptides, produced IFNg upon cocul-ture with gp100-expressing cell lines or tyrosinase-expres-sing cell lines (Table 2, Fig. 4C), suggesting that TAA-specificT cells with another specificity than the epitopes used forpeptide stimulation and tetramer staining were induced bythe DC vaccine.

Figure 1. KLH-specific immune responses before and after DC vaccination. A, KLH-specific T-cell proliferation was analyzed before the first vaccination andafter each DC vaccination during the first vaccination cycle in PBMC of stage III (filled circles) and stage IV (open circles) melanoma patients. Pertime point each dot represents 1 patient. Proliferative response to KLH is given as proliferation index (proliferationwith KLH/proliferationw/o KLH). �,P < 0.05;NS, not significant. B, KLH-specific IgG, IgA, and IgM antibodies were quantitatively measured after the first vaccination cycle in sera of vaccinated patients.Numbers indicate the number of patients without proliferative (A) or humoral (B) KLH-response.

Figure 2. Tumor antigen-specific CD8þ T-cell responses in peripheralblood. An example of tetramer analysis of PBMCs from patient II-E-08 isshown. Cells were stained with tetramers encompassing the HLA�02-specific gp100:154–168, gp100:280 to 288, tyrosinase:369 to 377peptide or an irrelevant peptide and with anti-CD8 mAb. The irrelevantcontrol peptide stained 0.01% of the PBMCs.

Aarntzen et al.

Clin Cancer Res; 18(19) October 1, 2012 Clinical Cancer ResearchOF6

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 7: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

Table 2. Immunologic and clinical responses

Flu-likesymptoms

Injectionsite reaction

TAA-specific T cellsin blood,

prevaccinationa

TAA-specific T cellsin blood,

postvaccinationa TAA-specific T cells in SKIL cultures

PatientCTCgrade

CTCgrade

CD4þ

TetramerþCD8þ

TetramerþCD4þ

TetramerþCD8þ

TetramerþCD8þ

Tetramerþa Peptideb Proteinb TumorbClinicalresponseg

PFS(mo)

OS(mo)

Stage IVII-E-01 1 0 n.a. – n.a. – – – – – PD 3 11IV-C-01 0 0 – – þþ – þþþ – – – SD 6 14IV-C-02 1 0 – – þþ – þ – þþ þ MR 5 10IV-C-05 1 1 – – – þ þþ – – – SD 16d 49d

IV-C-06 0 0 – – – – – – – – PD 1 3IV-C-09 1 2 – – – – – – – – PD 3 15IV-C-13 0 0 – – – – – – – – PD 1 5IV-D-01 0 0 n.a. – n.a. – þþ – – – PD 3 5IV-D-02 1 0 n.a. – n.a. – þ – – – PD 2 5IV-D-03 0 0 n.a. – n.a. þ – – – – SD 6 26IV-D-04 1 1 n.a. – n.a. – – – – – PD 3 22IV-D-06 2 1 n.a. þ n.a. þ þ þ þ – SD 16 24IV-D-09 1 0 n.a. – n.a. – – – – – SD 7 14IV-D-11 1 1 n.a. – n.a. – þþþ þ þ þ PR 8 52þIV-D-15 0 0 n.a. – n.a. – þ – – – PD 1e 8e

IV-D-17 0 0 n.a. – n.a. – – – – – PD 2 2IV-D-18 2 1 n.a. – n.a. – þ – – – SD 10 22IV-D-20 0 0 n.a. – n.a. – þþ – – – PD 4 21IV-D-21 1 0 n.a. – n.a. – þ – – – PD 1 7Stage IIIII-E-02 0 0 n.a. – n.a. – – – – – NED 34 54þII-E-03 2 1 – – – – þþþ þþþ þþ þ NED 53þ 53þII-E-04 1 1 n.a. – n.a. – þþ þ þþ – NED 52þ 52þII-E-05 1 1 n.a. – n.a. – þ þ þþ – NED 37 52þII-E-06 2 0 n.a. – n.a. – þþþ þþþ þþ þ NED 14 17II-E-07 2 1 n.a. – n.a. – þþþ þþ þþ þ NED 51þ 51þII-E-08 1 0 n.a. þ n.a. þ þþþ þþþ þþ þ NED 9 50þII-E-09 0 1 n.a. – n.a. þ þþþ þþ þþ þ NED 24 25II-E-10 1 1 n.a. – n.a. – þþþ þ þ – NED 48þ 48þII-E-11 2 1 n.a. – n.a. – þþ – – – NED 47þ 47þIV-C-03 2 1 –

f– þ – þþ þ þ þ NED 21 30

IV-C-04 1 0 – – – – – – – – NED 8 22IV-C-07 1 1 – – – – þþþ þþ þ þ NED 46þ 46þIV-C-08 2 1 – – þ – – – – – NED 40þ 40þIV-C-10 2 1 – – – – þþþ – þþ – NED 37þ 37þIV-C-11 1 1 – – – – – – – – NED 36þ 36þIV-C-12 1 1 – þ – þ þ þ þþ þ NED 8 16IV-C-14 1 0 – – – – –

c– þ þ NED 15 28þ

IV-D-05 1 1 n.a. – n.a. – þþ þþ þ þ NED 8 16IV-D-08 2 1 n.a. – n.a. – – – – – NED 10 12IV-D-10 0 1 n.a. – n.a. – þþ þþþ – – NED 23 28IV-D-14 0 0 n.a. – n.a. – þ þ – – NED 51þ 51þIV-D-16 0 1 n.a. – n.a. – – – – – NED 50þ 50þIV-D-19 1 1 n.a. – n.a. – þþ þ þ – NED 26 48þIV-D-22 0 0 n.a. – n.a. – – – – – PD 3 47þIV-D-24 0 0 n.a. – n.a. – – – – – NED 36þ 36þ

Abbreviations: n.a., not applicable; n.t., not tested; OS, overall survival; PFS, progression-free survival; SKIL, skin-test infiltrating lymphocytes.aTetramer staining of freshly isolated PBMC or SKILs. �, no recognition; þ, 1 epitope recognized; þþ, 2 epitopes recognized; þþþ, 3 epitopesrecognized.bAntigen recognition of SKILs after stimulation with T2 cells loaded with HLA-A2.1-binding gp100 or tyrosinase peptides (peptide recognition), BLMtransfected with gp100 or tyrosinase protein (protein recognition) or the gp100 and tyrosinase-expressing tumor cell line Mel624 (tumor recognition) asanalyzed by IFNg production. Responses were scored as the best immunologic response after 1 to 3 cycles of DC vaccinations.cPatient IV-C-14 had functional T cells without recognizing the tested epitopes (see Fig. 4).dNot evaluable for clinical response because no target lesion at start of vaccination.eNot evaluable for clinical response because of symptomatic brain metastases during 1st cycle.fgp100-specific CD4þ T cells were found after in vitro restimulation with DR4-binding peptides.gSD, stable disease; PD, progressive disease; NED, no evidence of disease; PR, partial response; MR, mixed response.

Vaccination of Melanoma Patients with mRNA-Electroporated DC

www.aacrjournals.org Clin Cancer Res; 18(19) October 1, 2012 OF7

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 8: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

Clinical outcome in stage III patientsOne patient had progressive disease within 4 months

after start of vaccinations. As of June 2011, 15 out of 26patients progressed at 3 to 37 months after the start ofvaccination. Twelve of 26 patients are in ongoing remissionfor up to 45 months. The median progression-free survival(PFS) is 34.3 months, and the median overall survival hasnot yet been reached. Extended follow-up is necessary todraw conclusions on thepotency of vaccinationwithmRNAelectroporated DCs as an adjuvant therapy in melanomapatients.

Clinical responses in stage IV patientsAll stage IV patientswere evaluated for clinical response at

3-month intervals with CT scan. Five patients had stabledisease up to 15 months and 1 patient (IV-C-02) showed amixed response. One patient (IV-D-11) with irresectableprimary melanoma of the nasal mucosa with bilaterallymph node metastases in the neck region and metastasesin the maxillary sinus, showed a partial response of theprimary tumor after 3 vaccinations, allowing resection ofthe primary tumor. The lymph node metastases and sinusmetastases completely regressed upon further vaccinationand this patient is in ongoing complete remission for 52þ

months (Fig. 5). DR4 expression was not correlated withsurvival (data not shown) in the vaccinated patients.

We observed a trend toward improved PFS in patientswith TAA-specific T cells in their blood or SKIL culturescompared with patients without TAA-specific responses,with 8.1 month versus 2.8 months, respectively (P ¼0.062). Similarly, patients with TAA-specific T cells showedimproved overall survival compared with patients withoutTAA-specific T cells, 24.1 months versus 11.0 months,respectively (P ¼ 0.101).

DiscussionEarly clinical trials have shown that vaccination with DC

loaded with tumor peptides is feasible, safe, and can inducetumor-specific immune responses in advanced cancerpatients (1, 5, 24). Although these results are promising,further improvement is warranted before its use can beaccepted in clinical practice. In the present study, we showthat DC presenting multiple naturally processed epitopesfollowing mRNA electroporation, enhance tumor-specificCD8þ and CD4þ T cell responses in melanoma patients.Importantly, both the presence of TAA-specific CD8þ T cellsand their capacity to produce IFNg upon encounter of theircognate antigen was significantly increased in stage IIIpatients treated in the adjuvant setting.

Long-lasting T-cell receptor stimulation of several hoursby fully matured DCs is necessary to activate naive T cells toproliferate and differentiate into effector cells (25, 26). Thegenerated DCs highly and sustainably expressed gp100 andtyrosinase after electroporation with mRNA. In vitro, DCwere able to activate gp100-specific CTL up to 48 hours afterelectroporation. Previously, we showed that gp100 andtyrosinase protein can be detected inside lymph nodes upto 24 hours after intranodal injection of mRNA electropo-rated DC (10). In this study, TAA-specific T cells wereinduced in the majority of patients, which clearly showsthat electroporated DC are indeed potent inducers oftumor-specific T cells.

We detected TAA-specific CD8þ T cells in peripheralblood of only 6 of the 45 patients. This is an underestima-tion, likely because of the low frequencies of these cells inthe circulation and the observation that substantially moreTAA-specific CD8þ T cells were detected in SKIL cultures.Still, the number of TAA-specific T cells measured in thisstudy might be underestimated because we screened withHLA�02:01 binding tetramers only. Indeed, in 6 patientsSKILs produced IFNg upon coculture with the proteingp100 and/or tyrosinase, although no IFNg productionwasdetected upon coculture with the corresponding HLA�02-binding peptide(s), supporting the notion that T cells with abroader specificity than the HLA�02:01 epitopes wereinduced. Recently, Bonehill and colleagues reported on theuse of monocyte-derived DC electroporated with mRNAencoding multiple tumor antigens, CD40 ligand, activeTLR4, and CD70 (TriMix-DC; 27). Although they moni-tored tumor-specific T cells by using autologous Epstein–Barr virus-transformed B cells transfected with tumor

Figure 3. Tumor antigen-specific CD4þ T-cell responses in peripheralblood. An example of tetramer analysis of PBMCs of patient IV-C-02 afterthe first cycle ofDC vaccination is shown. A, freshly isolatedPBMCswerestained directly after isolation with tetramers encompassing the HLA-DR4-specific gp100:44–59 peptide, tyrosinase:448–462 peptide, or anirrelevant peptide and with anti-CD4 mAb. B, tetramer analysis of PBMCafter 8 days of in vitro restimulationwith DR4-binding gp100 or tyrosinasepeptides. Note that before restimulation (A), only gp100-specific CD4þ Tcells are found, whereas after restimulation (B) both gp100- andtyrosinase-specific CD4þ T cells are detectable. C, the in vitro stimulatedpopulation of gp100- or tyrosinase-specific CD4þ PBMCof patient IV-C-02 was further characterized for FoxP3 expression. TAA-specific CD4þ Tcells did not express FoxP3.

Aarntzen et al.

Clin Cancer Res; 18(19) October 1, 2012 Clinical Cancer ResearchOF8

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 9: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

antigens as target cells, comparable frequencies of gp100and tyrosinase-specific CD8þ T cells were found.In 4 out of 10 patients tested, mRNA-electroporated DC-

induced concomitant TAA-specific CD4þ T cells. The obser-vation that these cells did not express FoxP3, suggest thatthese cells were T helper cells and not regulatory T cells.Initially, the main function of CD4þ T helper 1 cells wasthought to be the production of cytokines providing growthand differentiation signals to precursor CTL to becomeeffector CTL (28). However, CD4þ T cells have also beenshown to participate in the elimination of tumor and themaintenance of long-term protective immunity (29–31). Inaddition, activated T helper cells can stimulate precursorCTLs by reciprocal activation of APCs, for instance viaCD40-CD40L interactions (32). Recently it was shown thatCD4þ T cells enhance the recruitment of CD8þ T cells to thelymph nodes (33) and tumor (34–36). Moreover, a directantitumor effect of T helper cells has been shown (37–39).This may be of particular relevance for the antitumorresponse against melanoma because this tumor type fre-quently expresses class II molecules constitutively (40, 41).

Indeed, CD4þ T-cell responses have been identified inperipheral blood from melanoma patients who remaineddisease-free after treatment of multiple relapses (39).

Our data suggests a trend toward improved overall sur-vival, when compared with recently reported survival datain comparative arms from large randomized prospectivestudies on immunotherapy with anti-CTLA4 antibodies inirresectable metastatic melanoma patients (42, 43). It istempting to speculate that the observed clinical responsesresult from vaccine-induced immune responses. Indeed,stage IV melanoma patients with TAA-specific T-cellresponses showed increased clinical outcome after vaccina-tion with mRNA-loaded DC when compared with patientswith no vaccine-enhanced TAA-specific T-cell responses.These data confirm and extend our previous findings thatthe presence of tumor-specific T cells in SKIL cultures iden-tifies a subgroup of patients that might benefit from immu-notherapy (21).Moreover, these studies show that sustaineddisease control can be achieved in increasing numbers ofpatients, but objective antitumor responses might takeseveral months to years to develop (44, 45). The, in general,

Figure 4. Tumor antigen-specific CD8þ T-cell responses in posttreatment SKIL cultures. The presence and functionality of TAA-specific T cells were tested inlymphocytes cultured form skin-test biopsies (SKILs). A, an example is shown of tetramer analysis of SKILs from patient II-E-07, cultured from a delayed-typehypersensitivity (DTH) reaction to DC pulsed with tumor peptides. Cells were stained with tetramers encompassing the gp100:154 peptide, gp100:280,tyrosinase or an irrelevant peptide (control), and with anti-CD8 mAb. The irrelevant control peptide stained 0.07% of the T cells. The biopsy contains gp100-and tyrosinase-specific CD8þ T cells. B, IFNg production by the same T cells after stimulation with T2 cells loaded with gp100:154–168 peptide andgp100:280–288 or tyrosinase:369–377 peptide (peptide stimulation), BLM cells expressing gp100 or tyrosinase protein (protein stimulation), or Mel624 cellsexpressing both gp100 and tyrosinase (tumor stimulation). C, example of functional responses of SKILs of patient IV-C-14, cultured from a biopsy of a DTHreaction to DC electroporated with tyrosinase mRNA, showing recognition of tyrosinase epitopes when presented by HLA-A2.1 positive tyrosinase-transfectedBLMcells or endogenously tyrosinaseexpressingMel624 cells, by the specificandelevatedproductionof IFNg , although it does not recognize thespecific epitopes used in previous vaccination studies. This indicates that a broad repertoire of TAA-specific T cells can be stimulated by vaccination withmRNA-transfected DC.

Vaccination of Melanoma Patients with mRNA-Electroporated DC

www.aacrjournals.org Clin Cancer Res; 18(19) October 1, 2012 OF9

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 10: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

delayed response patterns in immunotherapy and the high-response rates to novel targeted therapies in melanoma,obviouslywarrants future studies that combine bothmodal-ities to achieve durable tumor control. Such studies shouldimplement SKIL culture analyses pre- and postinterventioninbothactive and comparative arms to elucidate thedynam-ics and nature of the induced immune responses.

The higher tumor burden in stage IV as compared withstage III melanoma patients may hamper the induction ofeffective immune responses but instead favor local immunesuppression. The present study shows that robust immu-nologic responses are more frequently induced in patientswith no evidence of disease compared with patients withmacroscopic tumor burden. On the basis of the associationof tumor-specific T cells and improved clinical outcome,

this suggests that DC-based vaccination is a promisingadjuvant treatment of stage III melanoma patients. How-ever, extended follow-up is warranted to draw conclusionson the clinical efficacy of DC-based vaccination in this stageof disease.

In summary, the advantages of vaccination with DCelectroporated with mRNA encoding TAA include lack ofHLA-restriction, presentation of multiple TAA epitopes tobothCD8þ andCD4þT cells, and the subsequent inductionof a large repertoire of TAA-specific T cells. We show thatvaccination of melanoma patients with mRNA-electropo-rated DC induces robust tumor-specific CD4þ and CD8þ T-cell responses, in particular, in stage III melanoma patientstreated adjuvant to radical lymph node dissection.

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

Authors' ContributionsConception and design: D. Schuurhuis, I.J.M. de Vries, C.G. Figdor, C.J.A.Punt, G.J. AdemaDevelopment of methodology: W.J. Lesterhuis, J.F.M. Jacobs, D. Schuur-huis, K. Thielemans, I.J.M. de Vries, C.J.A. Punt, G.J. AdemaAcquisitionofdata (provided animals, acquired andmanagedpatients,provided facilities, etc.): E.H.J.G Aarntzen, G. Schreibelt, K. Bol, W.J.Lesterhuis, S. Croockewit, J.H.W. De Wilt, M.M. van Rossum, J.F.M. Jacobs,T. Duiveman-de Boer, R. Mus, I.J.M. de Vries, G.J. AdemaAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): E.H.J.G Aarntzen, G. Schreibelt, K. Bol, J.F.M. Jacobs, T. Duiveman-de Boer, I.J.M. de Vries, C.J.A. Punt, G.J. AdemaWriting, review, and/or revision of themanuscript: E.H.J.G Aarntzen, G.Schreibelt, K. Bol, S. Croockewit, W.A.M. Blokx, J.F.M. Jacobs, R. Mus, K.Thielemans, I.J.M. de Vries, C.G. Figdor, G.J. AdemaAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): E.H.J.G Aarntzen, G. Schreibelt, K.Bol, I.J.M. de Vries, G.J. AdemaStudy supervision: I.J.M. de Vries, C.J.A. Punt, G.J. Adema

AcknowledgmentsThe authors would like to thank Nicole Scharenborg, Annemiek de Boer,

Mandy van de Rakt, Michel OldeNordkamp, Christel van Riel, Inge Boullart,Marieke Kerkhoff, Jeanette Pots, Rian Bongaerts and Dr. S. Strijk for theirassistance.

Grant SupportThis work was supported by grants from the Dutch Cancer Society

(KUN2006-3699, KUN2009-4402), the EU (ENCITE HEALTH-F5-2008-201842, Cancer Immunotherapy LSHC-CT-2006-518234, and DC-THERALSB-CT-2004-512074), The Netherlands Organization for ScientificResearch (NWO-Vidi-917.76.363, AGIKO-92003250, AGIKO-2008-2-4),the NOTK foundation, and AGIKO RUNMC. C.G. Figdor received the NWOSpinoza award.

The costs of publicationof this articlewere defrayed inpart by thepaymentof page charges. This article must therefore be herebymarked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received February 23, 2012; revised July 5, 2012; accepted July 17, 2012;published OnlineFirst August 15, 2012.

References1. Lesterhuis WJ, Aarntzen EHJG, Vries IJM, Schuurhuis DH, Figdor CG,

Adema GJ, et al. Dendritic cell vaccines in melanoma: from promise toproof? Crit Rev Oncol Hematol 2008;66:118–34.

2. Lesterhuis WJ, de Vries IJ, Schreibelt G, Lambeck AJ, Aarntzen EH,Jacobs JF, et al. Route of administration modulates the induction ofdendritic cell vaccine-induced antigen-specific T cells in advancedmelanoma patients. Clin Cancer Res 2011;17:5725–35.

3. Schultz ES, Schuler-Thurner B, Stroobant V, Jenne L, Berger TG,Thielemanns K, et al. Functional analysis of tumor-specific Th cellresponses detected in melanoma patients after dendritic cell-basedimmunotherapy. J Immunol 2004;172:1304–10.

4. Verdijk P, Aarntzen EHJG, Lesterhuis WJ, Boullart ACI, Kok E, vanRossum MM, et al. Limited amounts of dendritic cells migrate intothe T-cell area of lymph nodes but have high immune activating

Figure 5. Partial response after 3 intranodal vaccinations with mRNA-transfected DC. A, patient IV-D-11 presented with a irresectable primarymelanoma from nasal mucosa with extension into the nasal septum,maxillary and ethmoid sinus, and bilateral lymphadenopathy of level 1, 2,and 5. B, 3 intranodal vaccinations with mRNA-transfected DC induce apartial response, allowing resection of the primary tumor. C, patient is inongoing remission.

Aarntzen et al.

Clin Cancer Res; 18(19) October 1, 2012 Clinical Cancer ResearchOF10

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 11: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

potential in melanoma patients. Clin Cancer Res 2009;15:2531–40.

5. Wilgenhof S, Van Nuffel AM, Corthals J, Heirman C, Tuyaerts S,Benteyn D, et al. Therapeutic vaccination with an autologous mRNAelectroporated dendritic cell vaccine in patients with advanced mel-anoma. J Immunother 2011;34:448–56.

6. Laverman P, de Vries IJM, Scharenborg NM, de Boer A, Broekema M,Oyen WJG, et al. Development of In-111-labeled tumor-associatedantigen peptides formonitoring dendritic-cell-based vaccination. NuclMed Biol 2006;33:453–8.

7. Mitchell DA, Nair SK. RNA-transfected dendritic cells in cancer immu-notherapy. J Clin Invest 2000;106:1065–9.

8. Skipper JCA, Hendrickson RC,Gulden PH, Brichard V, Vanpel A, ChenY, et al. An HLA-A2-restricted tyrosinase antigen on melanoma cellsresults from posttranslational modification and suggests a novelpathway for processing of membrane proteins. J Exp Med 1996;183:527–34.

9. Zarling AL, Ficarro SB, White FM, Shabanowitz J, Hunt DF, EngelhardVH. Phosphorylated peptides are naturally processed and presentedby major histocompatibility complex class I molecules in vivo. J ExpMed 2000;192:1755–62.

10. Schuurhuis DH, Verdijk P, Schreibelt G, Aarntzen EHJG, ScharenborgNM, deBoer A, et al. In situ expression of tumor antigens bymessengerRNA-electroporated dendritic cells in lymph nodes of melanomapatients. Cancer Res 2009;69:2927–34.

11. Van Tendeloo V, Ponsaerts P, Lardon F, Nijs G, Lenjou M, VanBroeckhoven C, et al. Highly efficient gene delivery by mRNA elec-troporation in humanhematopoietic cells: superiority to lipofection andpassive pulsing of mRNA and to electroporation of plasmid cDNA fortumor antigen loading of dendritic cells. Blood 2001;98:49–56.

12. Schuurhuis DH, Lesterhuis WJ, Kramer M, LoomanMGM, Hout-KuijerM, Schreibelt G, et al. Polyinosinic polycytidylic acid prevents efficientantigen expression after mRNA electroporation of clinical grade den-dritic cells. Cancer Immunol Immunother 2009;58:1109–15.

13. Gajewski TF, Meng Y, Harlin H. Immune suppression in the tumormicroenvironment. J Immunother 2006;29:233–40.

14. Gajewski TF. Failure at the effector phase: immune barriers at the levelof the melanoma tumor microenvironment. Clin Cancer Res 2007;13(18 Pt 1):5256–61.

15. Gajewski TF,MengY, BlankC, Brown I, Kacha A, Kline J, et al. Immuneresistance orchestrated by the tumormicroenvironment. Immunol Rev2006;213:131–45.

16. Eggermont AM, Testori A,Marsden J, Hersey P, Quirt I, Petrella T, et al.Utility of adjuvant systemic therapy in melanoma. Ann Oncol 2009;20(Suppl 6):vi30–4.

17. de Vries IJM, Lesterhuis WJ, Scharenborg NM, Engelen LPH, RuiterDJ, Gerritsen MJP, et al. Maturation of dendritic cells is a prerequisitefor inducing immune responses in advanced melanoma patients. ClinCancer Res 2003;9:5091–100.

18. Jonuleit H, Kuhn U, Muller G, Steinbrink K, Paragnik L, Schmitt E, et al.Proinflammatory cytokines and prostaglandins induce maturation ofpotent immunostimulatory dendritic cells under FCS-free conditions.Effect of culture conditions on the type of T cell response. EurJ Immunol 1997;27:3135–42.

19. de Vries IJM, Eggert AAO, Scharenborg NM, Vissers JLM, LesterhuisWJ, Boerman OC, et al. Phenotypical and functional characterizationof clinical grade dendritic cells. J Immunother 2002;25:429–38.

20. Figdor CG, de Vries IJM, Lesterhuis WJ, Melief CJM. Dendritic cellimmunotherapy: mapping the way. Nat Med 2004;10:475–80.

21. de Vries IJM, Bernsen MR, Lesterhuis WJ, Scharenborg NM, StrijkSP, Gerritsen MJP, et al. Immunomonitoring tumor-specific T cellsin delayed-type hypersensitivity skin biopsies after dendritic cellvaccination correlates with clinical outcome. J Clin Oncol 2005;23:5779–87.

22. Morse MA, Lyerly HK. Clinical applications of dendritic cell vaccines.Curr Opin Mol Ther 2000;2:20–8.

23. GermeauC,MaWB, Schiavetti F, LurquinC, Henry E, VigneronN, et al.High frequency of antitumor T cells in the blood of melanoma patientsbefore and after vaccination with tumor antigens. J Exp Med2005;201:241–8.

24. Kyte JA, Mu L, Aamdal S, Kvalheim G, Dueland S, Hauser M, et al.Phase I/II trial ofmelanoma therapywith dendritic cells transfectedwithautologous tumor-mRNA. Cancer Gene Ther 2006;13:905–18.

25. Hugues S, Fetler L, Bonifaz L, Helft J, Amblard F, Amigorena S. DistinctT cell dynamics in lymph nodes during the induction of tolerance andimmunity. Nat Immunol 2004;5:1235–42.

26. Iezzi G, Karjalainen K, Lanzavecchia A. The duration of antigenicstimulation determines the fate of naive and effector T cells. Immunity1998;8:89–95.

27. Bonehill A, Tuyaerts S, Van Nuffel AM, Heirman C, Bos TJ, Fostier K,et al. Enhancing theT-cell stimulatory capacity of humandendritic cellsby co-electroporation with CD40L, CD70 and constitutively activeTLR4 encoding mRNA. Mol Ther 2008;16:1170–80.

28. Keene JA, Forman J. Helper activity is required for the in vivo gener-ation of cytotoxic T lymphocytes. J Exp Med 1982;155:768–82.

29. HuHM,Winter H, UrbaWJ, Fox BA. Divergent roles for CD4þ T cells inthe priming and effector/memory phases of adoptive immunotherapy.J Immunol 2000;165:4246–53.

30. Ossendorp F, Mengede E, Camps M, Filius R, Melief CJM. Specific Thelper cell requirement for optimal induction of cytotoxic T lympho-cytes against major histocompatibility complex class II negativetumors. J Exp Med 1998;187:693–702.

31. Toes REM, Ossendorp F, Offringa R, Melief CJM. CD4 T cells and theirrole in antitumor immune responses. J Exp Med 1999;189:753–6.

32. Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40Linteractions. Nature 1998;393:480–3.

33. Kumamoto Y,Mattei LM, Sellers S, PayneGW, Iwasaki A. CD4þ T cellssupport cytotoxic T lymphocyte priming by controlling lymph nodeinput. Proc Natl Acad Sci U S A 2011;108:8749–54.

34. BosR, Sherman LA. CD4þ T-cell help in the tumormilieu is required forrecruitment and cytolytic function ofCD8þT lymphocytes. Cancer Res2010;70:8368–77.

35. Marzo AL, Kinnear BF, Lake RA, Frelinger JJ, Collins EJ, RobinsonBWS, et al. Tumor-specific CD4(þ) T cells have a major "post-licens-ing" role in CTL mediated anti-tumor immunity. J Immunol 2000;165:6047–55.

36. WongSB, Bos R, Sherman LA. Tumor-specificCD4þ T cells render thetumor environment permissive for infiltration by low-avidity CD8þ Tcells. J Immunol 2008;180:3122–31.

37. Baxevanis CN, Voutsas IF, Tsitsilonis OE, Gritzapis AD, Sotiriadou R,Papamichail M. Tumor-specific CD4þ T lymphocytes from cancerpatients are required for optimal induction of cytotoxic T cells againstthe autologous tumor. J Immunol 2000;164:3902–12.

38. Mellman I, Steinman RM. Dendritic cells: specialized and regulatedantigen processing machines. Cell 2001;106:255–8.

39. Takahashi T, Chapman PB, Yang SY, Hara I, Vijayasaradhi S,Houghton AN. Reactivity of autologous CD4þ T lymphocytes againsthumanmelanoma.Evidence for a sharedmelanomaantigenpresentedby HLA-DR15. J Immunol 1995;154:772–9.

40. Campillo JA, Martinez-Escribano JA, Muro M, Moya-Quiles R, MarinLA, Montes-Ares O, et al. HLA class I and class II frequencies inpatients with cutaneous malignant melanoma from southeasternSpain: the role of HLA-C in disease prognosis. Immunogenetics2006;57:926–33.

41. Rodriguez T, Mendez R, Del CA, Aptsiauri N, Martin J, Orozco G, et al.Patterns of constitutive and IFN-gamma inducible expression of HLAclass II molecules in human melanoma cell lines. Immunogenetics2007;59:123–33.

42. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, HaanenJB, et al. Improved survival with ipilimumab in patients with metastaticmelanoma. N Engl J Med 2010;363:711–23.

43. Robert C, Thomas L, Bondarenko I, O'Day S, JW MD, Garbe C, et al.Ipilimumab plus dacarbazine for previously untreated metastatic mel-anoma. N Engl J Med 2011;364:2517–26.

44. Hoos A, Eggermont AM, Janetzki S, Hodi FS, Ibrahim R, Anderson A,et al. Improved endpoints for cancer immunotherapy trials. J NatlCancer Inst 2010;102:1388–97.

45. Lesterhuis WJ, Haanen JB, Punt CJ. Cancer immunotherapy–revis-ited. Nat Rev Drug Discov 2011;10:591–600.

www.aacrjournals.org Clin Cancer Res; 18(19) October 1, 2012 OF11

Vaccination of Melanoma Patients with mRNA-Electroporated DC

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368

Page 12: Vaccination with mRNA-Electroporated Dendritic Cells · 2012. 9. 10. · Cancer Therapy: Clinical Vaccination with mRNA-Electroporated Dendritic Cells Induces Robust Tumor Antigen-Specific

Published OnlineFirst August 15, 2012.Clin Cancer Res   Erik H. J. G. Aarntzen, Gerty Schreibelt, Kalijn Bol, et al.   Cells Responses in Stage III and IV Melanoma Patients

T+ and CD8+Induces Robust Tumor Antigen-Specific CD4Vaccination with mRNA-Electroporated Dendritic Cells

  Updated version

  10.1158/1078-0432.CCR-11-3368doi:

Access the most recent version of this article at:

  Material

Supplementary

 

http://clincancerres.aacrjournals.org/content/suppl/2012/08/14/1078-0432.CCR-11-3368.DC1Access the most recent supplemental material at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://clincancerres.aacrjournals.org/content/early/2012/09/10/1078-0432.CCR-11-3368To request permission to re-use all or part of this article, use this link

Cancer Research. on November 24, 2020. © 2012 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst August 15, 2012; DOI: 10.1158/1078-0432.CCR-11-3368