telomerase peptide vaccination combined with temozolomide ... · lars julsrud2, and steinar...

14
Cancer Therapy: Clinical Telomerase Peptide Vaccination Combined with Temozolomide: A Clinical Trial in Stage IV Melanoma Patients Jon Amund Kyte 1,3 , Gustav Gaudernack 3,4 , Svein Dueland 1 , Sissel Trachsel 3 , Lars Julsrud 2 , and Steinar Aamdal 1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity, immune response, and clinical response in melanoma patients after combined therapy with temozolomide and the telomerase peptide vaccine GV1001. Our previous GV1001 trials showed immune responses in approximately 60% of lung or pancreatic cancer patients. Experimental Design: Twenty-five subjects with advanced stage IV melanoma (M1B or M1C) received concomitant temozolomide and GV1001. Temozolomide was administered 200 mg/m 2 orally for 5 days every fourth week, and GV1001 as eight injections over 11 weeks. Immune response was evaluated by delayed type hypersensitivity, T-cell proliferation, and cytokine assays. The immunologic responders continued monthly vaccination. Results: The treatment was well tolerated. A GV1001-specific immune response was shown in 18 of 23 evaluated subjects (78%). Patients developing long-term T-cell memory survived more than those rapidly losing their responses. The immune response exhibited several characteristics of possible clinical sig- nificance including high IFNg /IL-10 ratios, polyfunctional cytokine profiles, and recognition of naturally processed antigens. Survival compared favorably with matched controls from a benchmark meta-analysis (1 year: 44% vs. 24%, 2 years: 16% vs. 6.6%). The clinical responses developed gradually over years, contrary to what is expected from chemotherapy. Five patients developed partial tumor regression and six more recorded stable disease. One patient has no remaining disease on fluorodeoxyglucose positron emission tomography scans after 5 years. Conclusions: The immunologic response rate is considerable compared with previous GV1001 trials without concomitant chemotherapy, although low toxicity is retained. The results warrant further studies of GV1001/temozolomide treatment and support the general concept of combining cancer vaccination with chemotherapy. Clin Cancer Res; 17(13); 4568–80. Ó2011 AACR. Introduction Cancer chemotherapy had been to a large extent ineffec- tive until the development of broad regimes combining different agents. The limited efficacy of monotherapy reflects tumor heterogeneity and genetic instability, favor- ing escape of resistant tumor subpopulations. Cancer vac- cination offers a different angle of attack, as compared with cytostatics, and may therefore be particularly effective in combinatory regimes. However, most chemotherapeutic agents are immunosuppressive, and it was long assumed that vaccines should therefore not be combined with chemotherapy. More recent knowledge has challenged this conventional wisdom (1, 2). In particular, it is discussed whether chemotherapy may be utilized to counter tumor protection from regulatory T cells (Tregs), while prevailing the immunocompetence necessary for vaccine response (3–5). Here we report a proof-of-principle trial (phase I/II) of combined therapy with temozolomide and telomerase peptide vaccination in 25 stage IV melanoma patients. The primary objectives were toxicity and immunologic response. Tumor response was a secondary study objective. Current prognosis for metastatic melanoma is dismal, with median survival of 4 to 9 months in patients with visceral involvement (6, 7). Dacarbazine has been considered standard treatment, but the tumor response rate is only 6% to 15%, mostly partial responses, and there is no documented effect on survival (7). The oral drug temozo- lomide is metabolized to dacarbazine in vivo and carries a potential advantage in penetration of the blood–brain Authors' Affiliations: 1 Section for Clinical Cancer Research, Department of Oncology and 2 Department of Radiology, Oslo University Hospital; 3 Department of Immunology, Cancer Research Institute, Oslo University Hospital and University of Oslo; and 4 Faculty of Medicine, University of Oslo, Oslo, Norway Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Note: The study was presented in part as abstract at ASCO 2006 (abstract 8031). Corresponding Author: Jon Amund Kyte, Department of Oncology, Oslo University Hospital, Radiumhospitalet, 0310 Oslo, Norway. Phone: 47-22- 93-40-00; Fax: 47-22-93-58-08; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-0184 Ó2011 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 17(13) July 1, 2011 4568 Cancer Research. on September 29, 2020. © 2011 American Association for clincancerres.aacrjournals.org Downloaded from Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Upload: others

Post on 26-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

Cancer Therapy: Clinical

Telomerase Peptide Vaccination Combined with Temozolomide:A Clinical Trial in Stage IV Melanoma Patients

Jon Amund Kyte1,3, Gustav Gaudernack3,4, Svein Dueland1, Sissel Trachsel3,Lars Julsrud2, and Steinar Aamdal1,4

AbstractPurpose: The study is a proof-of-principle trial evaluating toxicity, immune response, and clinical

response in melanoma patients after combined therapy with temozolomide and the telomerase peptide

vaccine GV1001. Our previous GV1001 trials showed immune responses in approximately 60% of lung or

pancreatic cancer patients.

Experimental Design: Twenty-five subjects with advanced stage IV melanoma (M1B or M1C) received

concomitant temozolomide and GV1001. Temozolomide was administered 200 mg/m2 orally for 5 days

every fourth week, and GV1001 as eight injections over 11 weeks. Immune response was evaluated by

delayed type hypersensitivity, T-cell proliferation, and cytokine assays. The immunologic responders

continued monthly vaccination.

Results: The treatment was well tolerated. A GV1001-specific immune response was shown in 18 of 23

evaluated subjects (78%). Patients developing long-term T-cell memory survived more than those rapidly

losing their responses. The immune response exhibited several characteristics of possible clinical sig-

nificance including high IFNg/IL-10 ratios, polyfunctional cytokine profiles, and recognition of naturally

processed antigens. Survival compared favorably with matched controls from a benchmark meta-analysis

(1 year: 44% vs. 24%, 2 years: 16% vs. 6.6%). The clinical responses developed gradually over years,

contrary to what is expected from chemotherapy. Five patients developed partial tumor regression and six

more recorded stable disease. One patient has no remaining disease on fluorodeoxyglucose positron

emission tomography scans after 5 years.

Conclusions: The immunologic response rate is considerable compared with previous GV1001 trials

without concomitant chemotherapy, although low toxicity is retained. The results warrant further studies

of GV1001/temozolomide treatment and support the general concept of combining cancer vaccination

with chemotherapy. Clin Cancer Res; 17(13); 4568–80. �2011 AACR.

Introduction

Cancer chemotherapy had been to a large extent ineffec-tive until the development of broad regimes combiningdifferent agents. The limited efficacy of monotherapyreflects tumor heterogeneity and genetic instability, favor-ing escape of resistant tumor subpopulations. Cancer vac-cination offers a different angle of attack, as compared with

cytostatics, and may therefore be particularly effective incombinatory regimes. However, most chemotherapeuticagents are immunosuppressive, and it was long assumedthat vaccines should therefore not be combined withchemotherapy. More recent knowledge has challenged thisconventional wisdom (1, 2). In particular, it is discussedwhether chemotherapy may be utilized to counter tumorprotection from regulatory T cells (Tregs), while prevailingthe immunocompetence necessary for vaccine response(3–5).

Here we report a proof-of-principle trial (phase I/II) ofcombined therapy with temozolomide and telomerasepeptide vaccination in 25 stage IV melanoma patients.The primary objectives were toxicity and immunologicresponse. Tumor response was a secondary study objective.Current prognosis for metastatic melanoma is dismal, withmedian survival of 4 to 9 months in patients with visceralinvolvement (6, 7). Dacarbazine has been consideredstandard treatment, but the tumor response rate is only6% to 15%, mostly partial responses, and there is nodocumented effect on survival (7). The oral drug temozo-lomide is metabolized to dacarbazine in vivo and carries apotential advantage in penetration of the blood–brain

Authors' Affiliations: 1Section for Clinical Cancer Research, Departmentof Oncology and 2Department of Radiology, Oslo University Hospital;3Department of Immunology, Cancer Research Institute, Oslo UniversityHospital and University of Oslo; and 4Faculty of Medicine, University ofOslo, Oslo, Norway

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

Note: The study was presented in part as abstract at ASCO 2006 (abstract8031).

Corresponding Author: Jon Amund Kyte, Department of Oncology, OsloUniversity Hospital, Radiumhospitalet, 0310 Oslo, Norway. Phone: 47-22-93-40-00; Fax: 47-22-93-58-08; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-0184

�2011 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 17(13) July 1, 20114568

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 2: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

barrier. In phase III trials, temozolomide and dacarbazinehave produced similar response rates (8, 9).Telomerase represents a possible target for universal

cancer vaccines (10, 11). The enzyme maintains telomerelength in dividing cells and is considered essential fortumor growth. Telomerase activity has been shown in allstudied cancer forms, including stem cell–like tumor cells(12–14). Interesting results have been reported fromphase I/II studies with 3 telomerase peptide vaccines(15–20). Peptide GV1001 represents a 16 amino acidsequence (EARPALLTSRLRFIPK) from the active site ofhuman telomerase reverse transcriptase (hTERT; refs. 21,22). The peptide was selected on the basis of computeralgorithms predicting strong human leukocyte antigen(HLA) class II binding properties and multiple nestedHLA class I binding motifs. The GV1001 vaccine may thusrecruit both CD4þ T helper cells and CD8þ cytotoxic Tcells. We have previously conducted 2 phase I/II trialswith GV1001, in pancreatic and lung cancer patients (19,20). Both studies were conducted without chemotherapy.The results indicated no serious adverse effects, a GV1001-specific immune response in approximately 60% ofpatients and an association between immune responseand survival. T-cell clone studies showed both T helperand cytotoxic responses (19, 20, 23). In the present trial,we hypothesized that temozolomide may enhance theimmune response by inducing antigen release or mod-ulating tumor tolerance. The study, to our knowledge, isthe first clinical trial evaluating combinatory therapy withtemozolomide and a cancer vaccine.

Patients and Methods

PatientsTwenty-five subjects with stage IV melanoma were

enrolled between January 2004 andNovember 2005. Inclu-sion criteria: histologically confirmed melanoma, nonre-sectable metastatic disease (AJCC stage IV), any HLAgenotype, measurable tumor, age between 18 and 75 years,Eastern Cooperative Oncology Group (ECOG) perfor-mance status 0–2, adequate hematologic, renal, and hepaticfunction. Exclusion criteria: previous chemotherapy, clin-ical signs of brain metastases, severe cardiac disease, severeactive infections, and need for immunosuppressivemedica-tion. Screening for brain metastases was not done.

The trial was approved by the Norwegian MedicinesAgency, the Regional Committee for Medical ResearchEthics, and the Hospital Review Board. It was carried outin compliance with theWorldMedical AssociationDeclara-tion of Helsinki. Written informed consent was obtainedfrom all patients.

TreatmentThe treatment schedule is shown in Appendix Figure A1

(see supplementary data online). Temozolomide wasadministered according to standard recommendationsfor malignant melanoma, 200 mg/m2 orally on 5 conse-cutive days every 28 days. GV1001 was given as 3 injectionsduring week 2 (Monday, Wednesday, Friday), 2 injectionsduring week 3 (Monday, Friday), and single injections atweeks 6, 7, and 11. Immune responders were given boostervaccines at 4-week intervals. GV1001 (300 nmol peptide in125 mL saline) and adjuvant granulocyte macrophage col-ony stimulating factor (GM-CSF; 75 mg) was administeredby intradermal injection in the right paraumbilical area.

The duration of treatment depended on the clinical andimmunologic response, according to the protocol: patientswith objective tumor response or stable disease (SD) atweek 12 and immunologic response received additionalcycles with temozolomide and GV1001, with responseevaluation at every third cycle. Patients with objectiveresponse/SDwithout immunologic response received addi-tional cycles with temozolomide only. Patients with pro-gressive disease (PD) and immunologic responsediscontinued temozolomide, but were offered boostervaccination with GV1001 every 4 weeks. All patients com-pleting the first 7-week sequence of vaccines (23 of 25patients) were considered evaluable for immunologicresponse.

Clinical evaluationTumor response was assessed by computed tomography

(CT) scans and clinical examination at 12-week intervals,and classified according to the Response Evaluation Cri-teria in Solid Tumors (RECIST; ref. 24). Tumor responseand survival was evaluated by intention-to-treat analysis.

Blood screening and assessment of adverse drug reac-tions were conducted at each visit. Adverse events weregraded according to the National Cancer Institute common

Translational Relevance

Cancer vaccination and chemotherapy attack thetumor from different angels and may synergize to over-come tumor escape. Chemotherapy may facilitateimmunization by provoking inflammation and elimi-nating regulatory T cells. The present trial evaluatedcombined therapy with temozolomide and the telomer-ase peptide vaccine GV1001 in advanced melanomapatients. The results showed that 18 of 23 subjectsdeveloped a specific immune response, without notabletoxicity. Patients developing long-term immunologicmemory survived more than those rapidly loosing theirresponses. Survival was extended compared withmatched controls from a benchmark meta-analysis (1year: 44% vs. 24%). Five patients developed partialtumor regression, including 1 survivor that is negativeon positron emission tomography–computed tomogra-phy scans after 5 years. The 78% immune response rateis considerable compared with most vaccine trials,including previous GV1001 studies without concomi-tant chemotherapy. The results warrant further studiesof GV1001/temozolomide treatment and supportthe concept of combining cancer vaccination withchemotherapy.

Telomerase Vaccine and Temozolomide in Stage IV Melanoma

www.aacrjournals.org Clin Cancer Res; 17(13) July 1, 2011 4569

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 3: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

toxicity criteria (NCI-CTC) version 3.0 and consideredrelated to treatment if the relationship was reported asprobable or suspected.

Immunologic evaluationPeripheral blood mononuclear cells (PBMC) were

obtained at weeks 0, 5, 9, and 12 and at every third boostervaccination. Pre- and postvaccination samples were ana-lyzed in parallel for proliferation response to GV1001peptide stimulation (3H-Thymidine assays). T-cellresponses were considered GV1001-specific when the sti-mulatory index (SI; response with antigen divided byresponse without antigen) was more than 2. Specificimmune responses were further characterized by cytokineassays and T-cell clone experiments (HLA restriction, spe-cificity for truncated peptides, response to recombinanthTERT protein). Materials and methods for immunologicassays, delayed type hypersensitivity (DTH) recording, flowcytometry, and production of peptides and recombinanthTERT protein are described in Appendix 1 (see supple-mentary data online).

StatisticsThe study reports overall survival (OS) and progression-

free survival (PFS), calculated from start of study treatment.OS for the intention-to-treat population, calculated by theKaplan–Meier method, was compared with predicted sur-vival based on a meta-analysis of clinical trials including2,100 patients (6). The meta-analysis was conducted byKorn and colleagues for providing survival benchmarks forsmall scale trials. We carried out the calculation as recom-mended (6). Briefly, prognostic factors (sex, ECOG status,metastatic category) from our patients were inserted into amathematical formula generating predicted survival. Thecalculation was based on trials where brainmetastases werenot excluded, because we did not conduct screening forbrain involvement (see Discussion). A 95% CI was gener-ated for survival of the study group (n ¼ 25), whereas thepredicted survival curve was regarded as a fixed estimate(Fig. 3). Survival after 1 year for the study group wascompared with predicted survival by use of Student’s t test.

Results

Patient characteristicsTwenty-five patients with advanced stage IV melanoma

were enrolled (Table 1). The majority (16 of 25) had M1Cdisease (visceral metastases or elevated lactate dehydrogen-ase); the remaining 9 patients hadM1B disease (pulmonarymetastases). Metastatic sites, age, sex, and treatment detailsare listed in Appendix Table A1 (see supplementary dataonline).

ToxicityThe combined therapy with temozolomide and

GV1001 was well tolerated. One patient developed tran-sient neutropenia (CTC grade IV) and thrombocytopenia(grade III), considered related to temozolomide. Other-

wise, no treatment-related grade III/IV toxicity wasobserved. Milder side effects were recorded in all patients,most commonly representing nausea, vomiting, fatigue,or local skin reactions at the injection site. Allergic reac-tions (grade II) were observed in 2 subjects. Finally, therewas no evidence of long-term toxicity in follow-up sam-ples from patients with extended survival, in spite ofdurable GV1001-specific responses and repeated boostervaccination (maximum observation 5 years, see the fol-lowing text).

Induction of immune responsesT-cell proliferation assays were carried out on pre- and

postvaccination PBMCs from 23 of 25 patients, that is, inall patients whose relevant PBMCs were obtained. AGV1001-specific T-cell response was shown in 18 of 23(78%) of patients after vaccination (Fig. 1A). Six subjectshad a GV1001-response prior to vaccination (Fig. 1A),suggesting spontaneous priming against naturallyexpressed GV1001 or cross-reacting epitopes. The SIincreased substantially after vaccination even in these 6patients (2.1!79; 3.2!35; 4.6!29; 7.5!24; 11!116;26!122). Among those developing de novo responses,40% developed a response by week 5 and 90% by week 9.

The DTH reactions were negative during standard studytreatment in all subjects whose recordings were obtained,including 11 immune responders in T-cell assays. Interest-ingly, 3 patients turned positive after omission of temozo-lomide and continuous booster vaccination. In ourprevious GV1001 trials without temozolomide (19, 20),the DTH reactions were positive in a substantial number ofpatients (26 of 62). These observations may point to amodulating effect of temozolomide on the GV1001response. DTH reactions have been associated with Th-1profiles, but cytokine analyses showed that the presentDTH negativity did not reflect low-level Th1 cytokines(see the following text).

Development of long-term T-cell memoryMost cancer vaccine trials have for practical reasons been

limited to short-term vaccination and immunomonitoring,whereas clinical efficacy probably requires long-term T-cellresponses (25, 26). Here, immune responders were offeredcontinued monthly vaccination. Long-term follow-uprevealed that 10 of 12 patients continuing vaccinationdeveloped durable GV1001-specific T-cell activity. Strik-ingly, all the 10 patients with durable T-cell responses atmonth 6 survived more than the 2 patients rapidly losingtheir responses (Table 1). Moreover, the survivors exhibitedretained responses in follow-up samples obtained at latertime points, ranging from week 36 to week 258 (Fig. 1B;Table 1; data not shown).

Tumor responseAll 25 patients enrolled had PD at study entry and

belonged to the most advanced categories within stageIV (64% M1C; 36% M1B; 0% M1A). Interestingly, 5 sub-jects developed partial tumor regression (partial response;

Kyte et al.

Clin Cancer Res; 17(13) July 1, 2011 Clinical Cancer Research4570

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 4: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

PR). Their tumor volumes decreased by 65% to 96%(Fig. 2A). An additional 6 patients experienced diseasestabilization, whereas 14 subjects had continued PD. All5 patients with PR developed GV1001-specific T-cellresponses, and the immune responses prevailed through-out their tumor regression periods (Fig. 1B). Interestingly,the clinical responses developed far more gradually thanexpected from chemotherapy, with only 1 reaching PR atweek 12, 3 more within week 36, and the fifth at week 48.Patient 19 (P19) had disseminated MIC melanoma at

study entry, including multiple visceral metastases. After

start of vaccination, no new lesions appeared. The totaltumor diameter started decreasing gradually, reached PR atweek 48, and has continued to regress over a periodexceeding 5 years (Fig. 2B). Remarkably, the patient hasno symptoms of tumor disease, and all residual CT-lesionsare negative on fluorodeoxyglucose positron emissiontomography (FDG-PET) scans. As melanomas are highlyPET-sensitive, the patient may have a complete response(CR). She currently receives booster vaccines at 3-monthintervals and retains a strong GV1001-specific immuneresponse (Fig. 1B).

Table 1. Disease stage, immune response, and clinical outcome

Patient AJCCstagea

T-cellresponseb

SI aftervaccc

Long-termT-cell memoryd

Tumorresponsee

PFS(days)f

Survival (days)g

P1 IV; M1C NE NE NA PD - 68P2 IV; M1C POS 14 POS (>6 mo) SD 168 344P3 IV; M1C POS 11 NA PD - 257P4 IV; M1C NE NE NA PD - 84P5 IV; M1C POS 122 NA PD - 583P6 IV; M1B POS 35 POS (>9 mo) SD 168 379P7 IV; M1B POS 26 POS (>9 mo) PD - 317P8 IV; M1C POS 3.7 NEG (<6 mo) SD 140 289P9 IV; M1C POS 39 NEG (<6 mo) PD - 283P10 IV; M1C POS 12 NA PD - 194P11 IV; M1B POS 11 POS (>25 mo) PR 585 1,134P12 IV; M1B POS 8.0 NA PD - 151P13 IV; M1B POS 79 POS (>14 mo) PR 502 563P14 IV; M1C NEG 0.8 NA PD - 99P15 IV; M1C NEG 1.3 NA PD - 250P16 IV; M1C POS 15 POS (>10 mo) PR 385 795P17 IV; M1B NEG 1.3 NA PD - 1,998þ (alive)h

P18 IV; M1C POS 17 NA PD - 239P19 IV; M1C POS 61 POS (>60 mo) PRi 1,905þ 1,905þ (alive)P20 IV; M1C POS 24 POS (>9 mo) SD 168 590P21 IV; M1B POS 29 POS (>13 mo) PD - 413P22 IV; M1C POS 122 POS (>14 mo) PR 248 594P23 IV; M1B NEG 1.1 NA SD 168 312P24 IV; M1C POS 116 NA PD - 418P25 IV; M1B NEG 1.1 NA SD 154 169

0 M1A 18/23 10/12 5 PR Mean: 4979 M1B 6 SD Median: 31716 M1C 14 PD

aDisease stage and metastasis category (within stage IV) at study entry.bPOS, positive, that is, T-cell proliferation specific for GV1001 (SI > 2); NEG, negative (no specific response); NE, not evaluable, that is,first round of vaccination (7 injections) not completed and postvaccination samples not available.cSI (response with GV1001 divided by response without GV1001) after vaccination (vacc).dLong-term T-cell response. POS, GV1001-specific response > 6 months (last available and positive sample is indicated in brackets);NEG, turning negative within 6 months; NA: not applicable (because no initial T-cell response or no available long-term samples).eTumor response (RECIST). Best response during vaccination (compared with baseline, evaluated at 12-week intervals).fPFS from start of study treatment. CT scan at study entry was used as reference.gSurvival from start of study treatment.hP17 was included in a different phase I study and developed clinical response.iP19 has some residual lesions on CT, but these lesions are negative on FDG-PET.

Telomerase Vaccine and Temozolomide in Stage IV Melanoma

www.aacrjournals.org Clin Cancer Res; 17(13) July 1, 2011 4571

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 5: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

0

00 5 9 12 24 36 48 60 73 83 97 108

25,000

Weeks

Weeks 10

0

10,000

20,000

30,000

40,000

50,000

25,000

50,000

75,000

100,000

125,000

5 9 12 24 44

0

0

20,000

40,000

60,000

80,000

cpm

cpm

PRPR

PR

SD

PR

PR

P22

0

30,000

60,000

90,000

120,000

150,000

0

10,000

20,000

30,000

cpm

40,000

50,000

5 9

PBMC + TPBMC + T + GV1001

PBMC + TPBMC + T + GV1001

PBMC + TPBMC + T + GV1001

PBMC + TPBMC + T + GV1001

PBMC + TPBMC + T + GV1001

PBMC + TPBMC + T + GV1001

P13P11

P16

P20

P19

12 24 36 48 60Weeks

0 5 9 12 24 36 48 60 72 85 97 109

121

132

144

156

169

192

206

219

232

245

258Weeks

Week 0 Week 5 Week 9 Week 12 Week 24 Week 36 Week 60Week 0 Week 5 Week 9 Week 12 Week 24 Week 39 Week 55

50,000

cpm

cpm

cpm

75,000

100,000

P2

P3

P5

P6

P7

P8

P9

P10

P11

Before vaccination

SI

After vaccination

P12

P13

P14

P15

P16

P17

P18

P19

P20

P21

P22

P23

P24

P25

5

10

15

20

25

30

35

40

45

50A

B

Figure 1. GV1001-specific T-cell responses. PBMCs were obtained prior to start of therapy, at weeks 5, 9, and 12 and at every third booster vaccination. ThePBMCs were stimulated once in vitro and tested for proliferation against irradiated PBMCs with or without peptide GV1001. A, pre- and postvaccinationT-cell response from all evaluated patients. Columns represent mean SI, that is, response with antigen (GV1001) divided by response without antigen.Responses with SI > 2 were considered GV1001-specific. B, long-term T-cell memory in clinical responders. Columns represent mean cpm of triplicates.Tumor response periods are indicated with horizontal bars below charts. The assays show durable GV1001-specific T-cell responses in 5 patients withPR and 1 with SD.

Kyte et al.

Clin Cancer Res; 17(13) July 1, 2011 Clinical Cancer Research4572

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 6: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

Figure 2. Tumor response. A, PRin 5 patients. Top, columnsrepresent total volume of allmeasurable lesions at study entryand minimum volume aftervaccination. Bottom, response ofindividual tumor lesions for eachpatient. Location of lesion: LH,liver hilus; LN, lymph node; Ma,mammary glands; Med,mediastnum; Mu, muscle; P,pulmonal; SC, subcutaneous Vvisceral. B; tumor response inpatient 19. Top, total diameter ofall measurable lesions(corresponding to RECIST) at timepoints for CT evaluation. P19stopped temozolomide at month8, but still receive boostervaccines at 3-month intervals.Middle, response of individuallesions at different time points. Allresidual CT lesions are negativeon FDG-PET scans, suggestingthat no viable tumor tissue mayremain. PET scans have not beenrepeated after month 42. Bottom,abdominal CT scans at study entryand after 61 months, showingregression of 3 metastatic lesions(CR ¼ complete regression; PR ¼partial regression). The month 61scan was taken without contrastdue to allergy.

0%P11

Ma

Starttherapy

00

3mStudy entry

Subcutan PR CR CR CR

CR

CR

PR CR

PRPR

PR

PR

CR

PR

PR

PR

PR

CR

PR

PR

PR

CR

PR

PR

PR

CR

PR

CR

PR

PR

PR

P

P

TumorTumor

Tumor

PR CR

CR KidneyKidney

Study entry Month 61

PR

Liver hilus

Pelvic

Visceral

Visceral

Visceral

Visceral

9m 12m 15m 31m 42m 61m

–1 3 6 9 12 14 17

Months from start therapy

P 1

9 cl

inic

al r

espo

nse

20 23 26 29 31 34 37 42 48 54 61

50

100

150

200

250

300

350

Tota

l tum

or d

ism

eter

(m

m)

Stoptemozol-omide

PETnegative

PP

P

P CR

CR

PRPR

PR

CR

CRCRCRCR

PRCRCRCRPR

SCLHVVVVV

CR PPP

MuSCSCLN

PRPRCR

CR

PRPR

PR

CRCR

PRPRPRCRPR

MedP

Lesions Lesions Lesions Lesions

Complete regression

Study entry

After therapy

Partial regression

Stable

Complete regressionPartial regression

Stable

Progression

PET negative

Lesions

PP

P

LNSCSCSC

LN

LNLN

P13 P16 P19 P22

20%

Tota

l tum

or v

olum

e(%

of b

asel

ine)

Indi

vidu

alta

rget

lesi

ons

40%

60%

80%

100%A

B

*

*

*

*

*

*

Telomerase Vaccine and Temozolomide in Stage IV Melanoma

www.aacrjournals.org Clin Cancer Res; 17(13) July 1, 2011 4573

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 7: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

Patient 11 (P11) had multiple metastases to lung andmammary glands at study entry. CT-evaluations showed a29% regression in total tumor diameter at week 12,improving to 74% at week 36. All 5 metastases regressedpartly or completely [Fig. 2A; Appendix Fig. A3 (see sup-plementary data online)]. We recorded PD at week 84 dueto a new subcutaneous lesion, but continued vaccinationand temozolomide. The patient stayed in good generalhealth for 3 years, before developing brain metastases.Subjects P13, P16, and P22 also responded with gradualtumor regression, accompanied by durable hTERT-specificT-cell responses (Figs. 1B and 2A). CT scans showed regres-sion of 6 of 8, 4 of 4, and 7 of 7 prestudy lesions,respectively [Fig. 2A; Appendix Figs. A2 and A3 (see sup-plementary data online)].

Progression-free and overall survivalTable 1 lists PFS and OS, calculated from start of study

treatment. By intention-to-treat analysis (n ¼ 25), medianOS was 10.4months (range, 2.2–66months) andmeanOS16.3 months (M1B 20 months; M1C 14months). Immuneresponders had increased median OS and PFS comparedwith nonresponders (median OS 396 days vs. 250 days;median PFS 112 days vs. 84 days). These differences did notreach statistical significance (log-rank test; P > 0.05), reflect-ing the low number of subjects. Advanced melanomapatients usually progress further within weeks or fewmonths (6, 8). It is therefore of interest that the 5 PRpatients exhibited extended PFS ranging from 8 to morethan 62 months, as well as OS ranging from 19 to morethan 62 months.

Trial subjectsmay differ fromother patients, for example,due to selection processes determining which individualsare referred to trial units. Korn and colleagues have con-ducted a meta-analysis of 2,100 trial patients and suggested

that survival in phase I/II melanoma studies is comparedwith their data set (6). They also outlined a method forcalculating predicted survival, correcting for prognosticfactors. Applying this method, we calculated a survivaladvantage for this study treatment, bordering95%statisticalsignificance (Fig. 3; intention-to-treat analysis). The appar-ent advantagewas relatively stable at different timepoints (1year: 44% vs. 24%; 2 years: 16% vs. 6.6%; 3 years: 12% vs.3.7%). Moreover, one may note that the "control group"was not based on untreated subjects, but on trial patientsreceiving potentially active therapy. Korn and colleaguessuggested declaring a treatment worthy of further study ifthe 1-year survival was better than predicted, with a P valueless than 0.10. This criterion was met both by intention-to-treat and per protocol analysis (P ¼ 0.059 and P ¼ 0.032,respectively). The interpretation of these findings is stillcomplex, as discussed in the following text.

T-cell clones from clinical respondersWe generated 8 GV1001-specific T-cell clones from 2

clinical responders (P11 and P22). The clones were HLAclass II DR restricted (Fig. 4A; data not shown). Takentogether with our data from other studies (refs. 19, 23;unpublished data), we find that GV1001 is recognized on aseries of DP, DR, and DQmolecules. The HLA promiscuityof GV1001 suggests that the vaccine is applicable to thegeneral patient population, without a need for HLA typing.

A diverse response in terms of HLA restriction and finespecificity may reduce the risk of tumor escape (25). Here,we determined the fine specificity of T-cell clones by experi-mentswith truncated peptides spanning theGV1001 aminoacid sequence. The 4 analyzed clones from subject P22recognized 3 different core motifs from the N-terminal partof GV1001 (Fig. 4B). One of themotifs was shared by a P11clone (Fig. 4D). In previous studies, we have observed thatmultiple motifs from the central and C-terminal parts ofpeptide GV1001 may also be recognized (21).

Vaccine-specific T-cell clones may be of sparse relevancein vivo if they fail to recognize naturally processed antigens.Here,weobserved recognitionofnaturally processedhTERTepitopes (Fig. 4C and D). Most clones responded morevigorously to hTERT protein than to pure GV1001 peptide(Fig. 4C; data not shown). We also noted strong responsesagainst low concentrations of truncated peptides (Fig. 4CandD). These observations suggest that the clonesmayhavehigher affinity for naturally processed epitopes than for thevaccine peptide. The data also point to a simplemechanismfor epitope spreading, as the cross-reacting natural epitopesmay stretch beyond the GV1001-sequence.

Cytokine profilesThe cytokine profiles for 14 of 18 immunologic respon-

ders were investigated in Bioplex assays by using panelsmeasuring up to 27 cytokines. The results showedGV1001-specific secretion of multiple cytokines in all14 patients [Fig. 5; Appendix Table A2 (see supplemen-tary data online)]. According to the Th1/Th2-paradigm, aTh1-like pattern is desirable for cancer eradication (27).

0.0

0 200 400 600 800 1,000

Days from start of study treatment

Sur

viva

l pro

port

ion

1,200 1,400

0.2

0.4

0.6

0.8

1.0Study cases (GV1001 + temozolomide; n = 25)

Controls (per Korn and colleagues data)

Figure 3. Survival in study patients compared with benchmark meta-analysis. Red line, OS for the study patients, calculated from start ofvaccination. Dashed red lines, 95% CI. Blue line, predicted survival for ourpatients (fixed estimate), calculated from Korn's meta-analysis ofmelanoma trials were brain metastases were not excluded (6). Thecalculation corrects for prognostic factors (performance status, site ofmetastases, etc.) and was done as recommended by Korn and colleagues.The analysis suggests a difference in favor of this study treatment,reaching a borderline statistical significance.

Kyte et al.

Clin Cancer Res; 17(13) July 1, 2011 Clinical Cancer Research4574

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 8: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

Figure 4. GV1001-specific T-cellclones from clinical responders.T-cell clone (T) proliferation afterstimulation with irradiated EBV-transformed cells (EB) with orwithout peptide GV1001 or hTERTprotein. Columns represent meancpm of triplicate wells. A, patient22. HLA restriction wasdetermined by blockage withmonoclonal antibodies againstDP, DQ, or DP molecules. B,patient 22. Fine specificityanalysis by stimulation withtruncated peptides covering theGV1001 sequence (amino acidsequences given in right text box).The 4 P22 clones recognize 3different core sequences,annotated in text boxes below thechart. C, patient 22. Recognitionof naturally processed antigens.T-cell clones were stimulated withEBV-transformed cells incubatedwith recombinant hTERT protein.The assay indicates response tonaturally processed protein inclones 5, 10, 18 (not 45). D, patient19. Fine specificity andrecognition of naturally processedantigens. T-cell clones werestimulated with truncatedpeptides or EBV-transformed cellsincubated with recombinanthTERT protein. The coresequence recognized ishighlighted in red below the chart.

0

0

0

0

2,000

4,000

cpm

6,000

8,000

10,000

15,000

30,000

45,000

60,000

75,000

90,000

40,000

80,000

120,000

160,000

#5 #10 #18

P22 clones: HLA restrictionA

B

C

D

P22 clones: Fine-specificities

P22 clones: hTERT protein reactivity

P11 clone 73:Fine specificity and hTERT protein reactivity

Core motif: EARPALLTSRLRFIPK

EB + TEB + T + GV1001

EB + T (no peptide)

EB + T + GV1001

GV1001

EB + T + GV1001 + aDREB + T + GV1001 + aDQEB + T + GV1001 + aDP

EB + T

EB + T + GV1001 (10 μmol/L)

GV1001 1 μmol/L

GV1001 100 nmol/L

hTERT protein 1.5 μmol/L

hTERT protein (6,5 μmol/L)hTERT protein (3,25 μmol/L)hTERT protein (1,625 μmol/L)

#45

#5EARPALLTS EARPALLTSR EARPALLTSR

EARPALLTSRLRFI

EARPALLTSRLRF

EARPALLTSRL

EARPALLTSR

EARPALLTS

ARPALLTSRLRFIPK

RPALLTSRLRFIPK

PALLTSRLRFIPK

ALLTSRLRFIPK

LLTSRLRFIPK

LTSRLRFIPK

TSRLRFIPK

PBMC + T (no peptide)GV1001 (EARPALLTSRLRFIPK)EARPALLTSRLRFIEARPALLTSRLRFEARPALLTSRLEARPALLTSREARPALLTSARPALLTSRLRFIPKRPALLTSRLRFIPKPALLTSRLRFIPKALLTSRLRFIPKLLTSRLRFIPKLTSRLRFIPKTSRLRFIPK

RPALLTSR#10 #18 #45

#5 #10 #18 #45

4000

8000

12,000

cpm

cpm

cpm

16,000

20,000

Telomerase Vaccine and Temozolomide in Stage IV Melanoma

www.aacrjournals.org Clin Cancer Res; 17(13) July 1, 2011 4575

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 9: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

0

1,000

2,000

pg/m

L

IFNγ TNFα IL-4

P2

PBMC + TPBMC + T + GV1001

P3 P6

P7P8 P9

P11 P13 P16

P19 P20 P21

P24P22

IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13 IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13IFNγ TNFα IL-4 IL-5 IL-10 IL-13

IFNγ TNFα IL-4 IL-5 IL-10 IL-13IFNγ TNFα IL-4 IL-5 IL-10 IL-13

3,000

4,000

0

1,000

2,000

pg/m

L

3,000

4,000

0

1,000

2,000pg/m

L

3,000

4,000

5,000

6,000

0

5,000

10,000

pg/m

L

15,000

20,000

25,000

30,000

0

1,000

2,000pg/m

L 3,000

4,000

5,000

0

1,000

2,000pg/m

L 3,000

4,000

5,000

0

500

1,000pg/m

L 1,500

2,000

2,500

0

50

100pg/m

L 150

200

250

0

50

100

pg/m

L

150

200

250

300

0

300

600

pg/m

L

900

0

50

100

150

pg/m

L

200

250

300

0

100

200

300pg/m

L 400

600

500

700

0

200

400

600

pg/m

L 800

1,200

1,000

1,400

0

200

400

pg/m

L

600

800

Figure 5. Secretion of Th1/Th2 cytokines in postvaccination T-cell cultures. Cytokine assays were carried out in 14 of 18 immune responders, that is, allsubjects were sufficient samples were obtained. T cells were stimulated with irradiated PBMCs with or without peptide GV1001. Supernatants wereanalyzed in duplicates or triplicates by Bioplex cytokine assays. Columns represent mean concentration (pg/mL). Data for non-Th1/Th2 cytokines are shown inAppendix Table A2 (see supplementary data online).

Kyte et al.

Clin Cancer Res; 17(13) July 1, 2011 Clinical Cancer Research4576

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 10: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

However, we have previously observed that humanresponses frequently do not follow a Th1/Th2 delineation(23, 28, 29). The present responses comprised high levelsof key Th1-cytokines IFNg and TNFa, but also of hallmarkTh2 cytokines, interleukin 5 (IL-5) and IL-13. Thisapplied both to the T-cell bulk cultures (Fig. 5) and tothe T-cell clones (ref. 23; data not shown). Collectively,our present and previous results indicate that cytokineprofiling should not rely on a Th1/Th2 dichotomy. Ofnote, we detected only low levels of IL-4 and IL-10(Fig. 5). The responses would therefore easily be desig-nated "Th1" with commonly used panels measuring onlyIFNg , TNFa, IL-4, and/or IL-10. Interestingly, the highIFNg/IL-10 ratios may reflect a favorable balance betweenimmunity and tolerance, as IL-10 is considered to pro-mote regulatory T cells and "tolerogenic" dendritic cells.The analyses further showed GV1001-specific secretion

of a broad range of proinflammatory cytokines and che-mokines [Appendix Table A2 (see supplementary dataonline)], including IL-1b, IL-6, IL-8, GM-CSF, and macro-phage inflammatory protein-1b. This polyfunctional cyto-kine profile was observed in all evaluated patients, both inT-cell bulk cultures and T-cell clones (Appendix Table A2(see supplementary data online); ref. 23; data not shown).We also noted that the GV1001-specific responses includedIL-17. A subset of activated T-cell clonesmay thus belong tothe Th17 lineage (30, 31).Finally, we asked whether temozolomide had affected

the cytokine profile of the GV1001 response and comparedthe cytokine patterns to data from patients in our otherGV1001 trials (Appendix Fig. A4 (see supplementary dataonline); refs. 20, 23; data not shown). The results suggestedno substantial difference in the range of cytokines secretedor the ratio between key cytokines.

Discussion

The concept of combining cancer vaccination with che-motherapy is attractive, and also challenging. Because ofthe long-standing assumption that chemotherapy wouldpreclude immunization, there is sparse experience on howthese modalities interact. Here, we combined temozolo-mide with an hTERT peptide vaccine. The lack of increasedtoxicity is of particular interest, as temozolomide is bonemarrow toxic and telomerase is expressed by hematologicstem cells. There is also no evidence of long-term toxicity insubjects receiving up to 5 years booster vaccination, in spiteof durable immune responses. Interestingly, the immuno-logic response rate of 78% is higher than that in anyprevious GV1001 trial (19–22, 32). Follow-up studies of12 immune responders revealed that 10 patients withdurable GV1001 responses all survived more than the 2subjects rapidly losing their T-cell activity. The clinicalevaluation showed objective tumor responses in 5 subjects.OS was extended compared with matched controls from abenchmark meta-analysis.The immune response rate of 78% indicates that temo-

zolomide did at least not preclude immunization. Most

subjects had normal lymphocyte counts at time of vaccina-tion (data not shown). This may be related to the temo-zolomide-free intervals in the dosage regime formelanoma. Temozolomide is known to affect T-cell counts,but mostly if administered continuously (33–36). Theconsiderable immune response rate may, moreover, reflecta beneficial effect of temozolomide. Several studies in micehave reported enhanced immunization after combiningvaccination with temozolomide, or that temozolomidesuppresses Treg function (4, 36–40). Chemotherapy mayalso suppress myeloid-derived suppressor cells (5).

The immunologic evaluation per protocol did notinclude Treg analyses, but we made an attempt to investi-gate the influence of temozolomide on Treg counts. Flowcytometry analyses showed that CD4þCD25high cells with aTreg phenotype were present in peripheral blood aftertemozolomide treatment and after the subsequent vaccineinjections (data not shown). We did not obtain sufficientpretreatment samples to determine whether temozolomidehad still affected Treg frequency and can therefore notmakeany conclusions about the effect of temozolomide on T-cellpopulations. In our ongoing melanoma vaccine trial(NCT00961844), this issue is addressed. Interestingly, pre-liminary data suggest that temozolomide inducesdecreased Treg counts (unpublished data). The cytokinedata reported earlier in the text indicated that temozolo-mide had not substantially altered the cytokine profilescompared with previous GV1001 trials. Nevertheless,temozolomide is known to induce apoptosis of melanomacells and is likely to induce inflammation and cross-prim-ing of tumor-associated antigens. We hypothesize that thisprocess may synergize with CD4þ T helper cells recruitedthrough GV1001 vaccination. T helper cells may in parti-cular engage antigen presenting cells (APC) presentingantigens from apoptotic tumor and induce epitope spread-ing (27). In ongoing studies of long-term survivors afterGV1001 vaccination, we have detected responses againsttelomerase antigens not included in the vaccine (unpub-lished data).

Although most peptide vaccines have represented shortCTL epitopes, the use of longer peptides recruiting T helpercells may yield advantages in vivo (1, 25, 27). A T helperresponse may more effectively synergize with apoptosisand inflammation induced by radiation or chemotherapy,as depicted earlier in the text. The durable memoryresponses reported herein for GV1001 point to anotherpossible advantage; several studies have suggested that Thelper activity is necessary for the development of CTLmemory (41, 42). Long peptides may, moreover, be pro-cessed by endogenous APCs and recruit T-cell clones withdiverse specificities, as observed for GV1001. Several longpeptides, including GV1001, have furthermore been shownto elicit cytotoxic responses against nested short epitopes(1, 19, 43–45).

The present GV1001 responders developed polyfunc-tional cytokine profiles with mixed Th1/Th2 patterns anda high IFNg/IL-10 ratio [Fig. 5; Appendix Table A2 (seesupplementary data online)]. The broad and proinflamma-

Telomerase Vaccine and Temozolomide in Stage IV Melanoma

www.aacrjournals.org Clin Cancer Res; 17(13) July 1, 2011 4577

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 11: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

tory cytokine profile may mobilize the adaptive and theinnate immune system. Interestingly, polyfunctional cyto-kine profiles have been associatedwith protective immunityin patients surviving HIV or Leishmania (46–48). Polyfunc-tional cytokine patterns may be particularly important in acancer vaccine setting, where there is a need to overcomeestablished tumor tolerance and transform the inflamma-tory milieu.

Six patients in the present trial harbored T-cell responsesagainst GV1001 prior to vaccination, and others havereported spontaneous GV1001-specific activity in subjectswith chronic lymphocytic leukemia (49). Furthermore, weshow that GV1001-specific T-cell clones recognize endo-genous APCs pulsed with hTERT protein. Both the pre-vaccination reactivity and the hTERT protein responsessuggest that GV1001-associated epitopes are naturally pro-cessed and immunogenic in cancer patients. This study didnot include screening of tumors for telomerase expressionand hence did not address whether quantitative differencesin expression level between patients may have influencedthe vaccine response.

We report objective tumor regression in 5 patients andtemporary disease stabilization in an additional 6 sub-jects. Several observations suggest that the regressions areunlikely to be caused only by temozolomide. First, allclinical responders belonged to the immunologicresponse group and developed long-term GV1001-speci-fic memory. Second, the response rate is relatively highcompared with the approximately 13% rate reported inphase III temozolomide trials (8, 50). Third, chemother-apy responses rarely occur in patients such as P19 withwidespread visceral metastases outside of lung. Fourth,P19 developed most of her tumor regression after omit-ting temozolomide at month 8 and continuing vaccina-tion (Fig. 2B). Fifth, responses to temozolomide arefrequently short-lived, whereas 4 of 5 responders hereexhibited PFS more than 1 year. Finally, responses tochemotherapy usually materialize shortly after start oftreatment. By contrast, 4 of 5 responses in the present trialfell short of PR criteria at month 3, and all responsesgradually improved over months or years. This pattern isin accordance with vaccine responses, depending onmobilizing the immune system and overcoming tumortolerance. Taken together, these observations suggest arole of GV1001 in the development of clinical responses.There is, however, a need to conduct a randomized studybefore making any conclusions on clinical efficacy. Theslow development of tumor responses shows that a con-ventional 3-month evaluation on the basis of RECISTmay be misleading in cancer vaccine trials and supportsthe concept of developing adjusted response criteria, assuggested by others (26).

The group of study patients survived more thanexpected, compared with predicted survival as calculatedfrom Korn’s meta-analysis. This finding should be inter-preted with utmost caution, but is in line with possibleclinical benefits suggested by the tumor regression andPFS data. Subjects with clinical signs of brain metastases

were excluded from our trial. On the basis of the highfrequency of asymptomatic brain involvement amongpatients with disseminated melanoma, it is still likely thata considerable fraction carried brain metastases. Becausescreening for cerebral involvement was not done, we calcu-lated predicted survival on the basis of Korn’s data fromstudies not excluding brain metastases. Of note, patientswith clinical signs of brain metastasis are rarely enrolled intrials, and the calculation takes into account the generallygoodperformance status in our patients. Survival in our trialwas calculated from the start of study treatment, whereasKorn’s meta-analysis gives survival from a possibly earlierpoint ("time of registration"). The latter difference suggeststhat our survival advantage may be underestimated. In all,these issues illustrate that any comparisonbetweendifferenttrials carries considerable uncertainty. The apparent survivalbenefit ofGV1001/temozolomide is interesting, but need tobe reproduced with a randomized control group.

We conclude that combined therapy with temozolomideand GV1001 is well tolerated in advanced melanomapatients, and that standard temozolomide dosage for mel-anoma does not preclude development of vaccineresponses. The immunologic responses exhibit several fea-tures of possible clinical significance, including durable T-cell memory, recognition of naturally processed antigens,high IFNg/IL-10 ratios andpolyfunctional cytokineprofiles.Collectively, present and previous trials show that peptideGV1001may recruit a diverse spectrumof T-cell clones withdifferent HLA restrictions and fine specificity. Regardingclinical outcome, we observed objective tumor responsesin 5 long-term immune responders. OS was extended com-pared with predicted survival from a meta-analysis. Therehave also been signs of a possible clinical activity in ourprevious GV1001 studies (19–21). However, both clinicaland immunologic response rates are higher in the presenttrial, whereas the low toxicity fromGV1001monotherapy isretained. In our opinion, the results warrant further studiesof combinatory therapy with GV1001 and temozolomide.Our findings also support the general concept of combiningcancer vaccination with chemotherapy.

Disclosure of Potential Conflicts of interest

G. Gaudernack is a member of the advisory board for Kael-Gemvax(patent holders for GV1001). The other authors declare no conflict ofinterest.

Acknowledgments

We thank M. Cvancarova for valuable help on statistical analysis, and J.A.Eriksen andM.Moeller at Gemvax.We also thank doctors and nurses at OsloUniversity Hospital for excellent clinical follow-up and patient care, andespecially K. Øwre, M. Nyakas, I. Sve, and L. Hegg. Gemvax AS, Norway,supplied GV1001 free of charge.

Grant Support

This work was supported by the Norwegian Cancer Society.

Received January 21, 2011; revised April 28, 2011; accepted May 8, 2011;published OnlineFirst May 17, 2011.

Kyte et al.

Clin Cancer Res; 17(13) July 1, 2011 Clinical Cancer Research4578

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 12: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

References1. Perez SA, von Hofe E, Kallinteris NL, Gritzapis AD, Peoples GE,

Papamichail M, et al. A new era in anticancer peptide vaccines.Cancer 2010;116:2071–80.

2. Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunologicalaspects of cancer chemotherapy. Nat Rev Immunol 2008;8:59–73.

3. Huang X, Huang G, Song H, Chen L. Preconditioning chemotherapywith paclitaxel and cisplatin enhances the antitumor activity of cyto-kine induced-killer cells in a murine lung carcinoma model. Int JCancer 2010. [Epub ahead of print].

4. Jordan JT, Sun W, Hussain SF, DeAngulo G, Prabhu SS, HeimbergerAB. Preferential migration of regulatory T cells mediated by glioma-secreted chemokines can be blocked with chemotherapy. CancerImmunol Immunother 2008;57:123–31.

5. Gonzalez-Aparicio M, Alzuguren P, Mauleon I, Medina-Echeverz J,Hervas-Stubbs S, Mancheno U, et al. Oxaliplatin in combination withliver-specific expression of interleukin 12 reduces the immunosup-pressive microenvironment of tumours and eradicates metastaticcolorectal cancer in mice. Gut 2010;60:341–49.

6. Korn EL, Liu PY, Lee SJ, Chapman JA, Niedzwiecki D, Suman VJ, et al.Meta-analysis of phase II cooperative group trials in metastatic stageIV melanoma to determine progression-free and overall survivalbenchmarks for future phase II trials. J Clin Oncol 2008;26:527–34.

7. Eigentler TK, Caroli UM, Radny P, Garbe C. Palliative therapy ofdisseminated malignant melanoma: a systematic review of 41 rando-mised clinical trials. Lancet Oncol 2003;4:748–59.

8. Middleton MR, Grob JJ, Aaronson N, Fierlbeck G, Tilgen W, Seiter S,et al. Randomized phase III study of temozolomide versus dacarba-zine in the treatment of patients with advanced metastatic malignantmelanoma. J Clin Oncol 2000;18:158–66.

9. Quirt I, Verma S, Petrella T, Bak K, Charette M. Temozolomide for thetreatment of metastatic melanoma: a systematic review. Oncologist2007;12:1114–23.

10. Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerasecatalytic subunit is a widely expressed tumor-associated antigenrecognized by cytotoxic T lymphocytes. Immunity 1999;10:673–9.

11. Harley CB. Telomerase and cancer therapeutics. Nat Rev Cancer2008;8:167–79.

12. Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D,et al. Isolation and in vitro propagation of tumorigenic breast cancercells with stem/progenitor cell properties. Cancer Res 2005;65:5506–11.

13. KimNW, PiatyszekMA, Prowse KR, Harley CB,West MD, Ho PL, et al.Specific association of human telomerase activity with immortal cellsand cancer. Science 1994;266:2011–5.

14. Shay JW, Bacchetti S. A survey of telomerase activity in humancancer. Eur J Cancer 1997;33:787–91.

15. Domchek SM, Recio A, Mick R, Clark CE, Carpenter EL, Fox KR, et al.Telomerase-specific T-cell immunity in breast cancer: effect ofvaccination on tumor immunosurveillance. Cancer Res 2007;67:10546–55.

16. Haining WN, Davies J, Kanzler H, Drury L, Brenn T, Evans J, et al. CpGoligodeoxynucleotides alter lymphocyte and dendritic cell traffickingin humans. Clin Cancer Res 2008;14:5626–34.

17. Bolonaki I, Kotsakis A, Papadimitraki E, Aggouraki D, Konsolakis G,Vagia A, et al. Vaccination of patients with advanced non-small-celllung cancer with an optimized cryptic human telomerase reversetranscriptase peptide. J Clin Oncol 2007;25:2727–34.

18. Parkhurst MR, Riley JP, Igarashi T, Li Y, Robbins PF, Rosenberg SA.Immunization of patients with the hTERT:540–548 peptide inducespeptide-reactive T lymphocytes that do not recognize tumors endo-genously expressing telomerase. Clin Cancer Res 2004;10:4688–98.

19. Brunsvig PF, Aamdal S, Gjertsen MK, Kvalheim G, Markowski-Grims-rud CJ, Sve I, et al. Telomerase peptide vaccination: a phase I/II studyin patients with non-small cell lung cancer. Cancer Immunol Immun-other 2006;55:1553–64.

20. Bernhardt SL, Gjertsen MK, Trachsel S, Møller M, Eriksen JA, Meo M,et al. Telomerase peptide vaccination of patients with non-resectablepancreatic cancer: A dose escalating phase I/II study. Br J Cancer2006;95:1474–82.

21. Kyte JA. Cancer vaccination with telomerase peptide GV1001. ExpertOpin Investig Drugs 2009;18:687–94.

22. Shaw VE, Naisbitt DJ, Costello E, Greenhalf W, Park BK, NeoptolemosJP, et al. Current status of GV1001 and other telomerase vaccinationstrategies in the treatment of cancer. Expert Rev Vaccines2010;9:1007–16.

23. Kyte JA, Trachsel S, Risberg B, thor Straten P, Lislerud K, GaudernackG. Unconventional cytokine profiles and development of T cell mem-ory in long-term survivors after cancer vaccination. Cancer ImmunolImmunother 2009;58:1609–26.

24. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS,Rubinstein L, et al. New guidelines to evaluate the response totreatment in solid tumors. J Natl Cancer Inst 2000;92:205–16.

25. Bijker MS, Melief CJ, Offringa R, Van Der Burg SH. Design anddevelopment of synthetic peptide vaccines: past, present and future.Expert Rev Vaccines 2007;6:591–603.

26. Wolchok JD, Hoos A, O’Day S, Weber JS, Hamid O, Lebb�e C, et al.Guidelines for the evaluation of immune therapy activity in solidtumors: immune-related response criteria. Clin Cancer Res 2009;15:7412–20.

27. Knutson KL, Disis ML. Tumor antigen-specific T helper cells in cancerimmunity and immunotherapy. Cancer Immunol Immunother2005;54:721–8.

28. Kyte JA, Mu L, Aamdal S, Kvalheim G, Dueland S, Hauser M, et al.Phase I/II trial of melanoma therapy with dendritic cells transfectedwith autologous tumor-mRNA. Cancer Gene Ther 2006;13:905–18.

29. Kyte JA, Kvalheim G, Lislerud K, thor Straten P, Dueland S, Aamdal S,et al. T cell responses in melanoma patients after vaccination withtumor-mRNA transfected dendritic cells. Cancer Immunol Immun-other 2007;56:659–75.

30. Dong C. Diversification of T-helper-cell lineages: finding the familyroot of IL-17-producing cells. Nat Rev Immunol 2006;6:329–33.

31. Romagnani S. Human Th17 cells. Arthritis Res Ther 2008;10:206–13.32. Greten TF, Forner A, Korangy F, N'Kontchou G, Barget N, Ayuso C,

et al. A phase II open label trial evaluating safety and efficacy of atelomerase peptide vaccination in patients with advanced hepatocel-lular carcinoma. BMC cancer 2010;10:209.

33. Brock CS, Newlands ES, Wedge SR, Bower M, Evans H, Colquhoun I,et al. Phase I trial of temozolomide using an extended continuous oralschedule. Cancer Res 1998;58:4363–7.

34. Tolcher AW, Gerson SL, Denis L, Geyer C, Hammond LA, Patnaik A,et al. Marked inactivation of O6-alkylguanine-DNA alkyltransferaseactivity with protracted temozolomide schedules. Br J Cancer2003;88:1004–11.

35. Wick W, Weller M. How lymphotoxic is dose-intensifiedtemozolomide? The glioblastoma experience. J Clin Oncol 2005;23:4235–6.

36. Su YB, Sohn S, Krown SE, Livingston PO, Wolchok JD, Quinn C, et al.Selective CD4þ lymphopenia in melanoma patients treated withtemozolomide: a toxicity with therapeutic implications. J Clin Oncol2004;22:610–6.

37. Kim CH,Woo SJ, Park JS, Kim HS, Park MY, Park SD, et al. Enhancedantitumour immunity by combined use of temozolomide and TAT-survivin pulsed dendritic cells in a murine glioma. Immunology2007;122:615–22.

38. Heimberger AB, Sun W, Hussain SF, Dey M, Crutcher L, Aldape K,et al. Immunological responses in a patient with glioblastoma multi-forme treated with sequential courses of temozolomide and immu-notherapy: case study. Neuro Oncol 2008;10:98–103.

39. Kim TG, Kim CH, Park JS, Park SD, Kim CK, Chung DS, et al.Immunological factors relating to the antitumor effect of temozolo-mide chemoimmunotherapy in a murine glioma model. Clin VaccineImmunol 2010;17:143–53.

40. Neyns B, Tosoni A, HwuWJ, Reardon DA. Dose-dense temozolomideregimens: antitumor activity, toxicity, and immunomodulatory effects.Cancer 116;2868–77.

41. Shedlock DJ, Shen H. Requirement for CD4 T cell help in generatingfunctional CD8 T cell memory. Science 2003;300:337–9.

Telomerase Vaccine and Temozolomide in Stage IV Melanoma

www.aacrjournals.org Clin Cancer Res; 17(13) July 1, 2011 4579

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 13: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

42. Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG,Schoenberger SP. CD4þ T cells are required for secondary expan-sion and memory in CD8þ T lymphocytes. Nature 2003;421:852–6.

43. Knutson KL, Schiffman K, Disis ML. Immunization with a HER-2/neuhelper peptide vaccine generates HER-2/neu CD8 T-cell immunity incancer patients. J Clin Invest 2001;107:477–84.

44. Gjertsen MK, Bjorheim J, Saeterdal I, Myklebust J, Gaudernack G.Cytotoxic CD4þ andCD8þT lymphocytes, generated bymutant p21-ras (12Val) peptide vaccination of a patient, recognize 12Val-depen-dent nested epitopes present within the vaccine peptide and killautologous tumour cells carrying this mutation. Int J Cancer1997;72:784–90.

45. Welters MJ, Kenter GG, Piersma SJ, Vloon AP, L€owik MJ, Berends-van der Meer DM, et al. Induction of tumor-specific CD4þ and CD8þT-cell immunity in cervical cancer patients by a human papillomavirustype 16 E6 and E7 long peptides vaccine. Clin Cancer Res2008;14:178–87.

46. Duvall MG, Precopio ML, Ambrozak DA, Jaye A, McMichael AJ,Whittle HC, et al. Polyfunctional T cell responses are a hallmark ofHIV-2 infection. Eur J Immunol 2008;38:350–63.

47. Betts MR, Nason MC, West SM, De Rosa SC, Migueles SA, AbrahamJ, et al. HIV nonprogressors preferentially maintain highly functionalHIV-specific CD8þ T cells. Blood 2006;107:4781–9.

48. Darrah PA, Patel DT, De Luca PM, Lindsay RW, Davey DF, Flynn BJ,et al. Multifunctional TH1 cells define a correlate of vaccine-mediatedprotection against Leishmania major. Nat Med 2007;13:843–50.

49. Kokhaei P, Palma M, Hansson L, Osterborg A, Mellstedt H, Choudh-ury A. Telomerase (hTERT 611–626) serves as a tumor antigen in B-cell chronic lymphocytic leukemia and generates spontaneouslyantileukemic, cytotoxic T cells. Exp Hematol 2007;35:297–304.

50. Kaufmann R, Spieth K, Leiter U, Mauch C, von den Driesch P, Vogt T,et al. Temozolomide in combination with interferon-alfa versus temo-zolomide alone in patients with advanced metastatic melanoma: arandomized, phase III, multicenter study from the Dermatologic Coop-erative Oncology Group. J Clin Oncol 2005;23:9001–7.

Kyte et al.

Clin Cancer Res; 17(13) July 1, 2011 Clinical Cancer Research4580

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184

Page 14: Telomerase Peptide Vaccination Combined with Temozolomide ... · Lars Julsrud2, and Steinar Aamdal1,4 Abstract Purpose: The study is a proof-of-principle trial evaluating toxicity,

2011;17:4568-4580. Published OnlineFirst May 17, 2011.Clin Cancer Res   Jon Amund Kyte, Gustav Gaudernack, Svein Dueland, et al.   PatientsTemozolomide: A Clinical Trial in Stage IV Melanoma Telomerase Peptide Vaccination Combined with

  Updated version

  10.1158/1078-0432.CCR-11-0184doi:

Access the most recent version of this article at:

  Material

Supplementary

 

http://clincancerres.aacrjournals.org/content/suppl/2011/05/17/1078-0432.CCR-11-0184.DC1Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/17/13/4568.full#ref-list-1

This article cites 48 articles, 18 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/17/13/4568.full#related-urls

This article has been cited by 9 HighWire-hosted articles. Access the articles at:

   

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

  SubscriptionsReprints 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/17/13/4568To request permission to re-use all or part of this article, use this link

Cancer Research. on September 29, 2020. © 2011 American Association forclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst May 17, 2011; DOI: 10.1158/1078-0432.CCR-11-0184