Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENT OF NON-SMALL CELL LUNG CARCINOMA BY ACTIVE IMMUNOTHERAPY
Document Type and Number:
WIPO Patent Application WO/2013/135266
Kind Code:
A1
Abstract:
Disclosed is a novel immunotherapeutic treatment regime for treatment of patients suffering from cancer, such as non-small cell lung cancer (NSCLC). The regime involves combination treatment with a peptide derived from human telomerase protein and GM-CSF in patients that have previously received chemoradiotherapy.

Inventors:
AAMDAL STEINAR (NO)
GAUDERNACK GUSTAV (NO)
Application Number:
PCT/EP2012/054272
Publication Date:
September 19, 2013
Filing Date:
March 12, 2012
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GEMVAX AS (NO)
AAMDAL STEINAR (NO)
GAUDERNACK GUSTAV (NO)
International Classes:
A61K38/10; A61P35/00
Domestic Patent References:
WO2011101173A12011-08-25
WO1998020027A21998-05-14
Other References:
BRUNSVIG PAAL FR ET AL: "Telomerase Peptide Vaccination in NSCLC: A Phase II Trial in Stage III Patients Vaccinated after Chemoradiotherapy and an 8-Year Update on a Phase I/II Trial", CLINICAL CANCER RESEARCH, vol. 17, no. 21, November 2011 (2011-11-01), pages 6847 - 6857, XP002678605
VONDERHEIDE R H ET AL: "THE TELOMERASE CATALYTIC SUBUNIT IS A WIDELY EXPRESSED TUMOR- ASSOCIATED ANTIGEN RECOGNIZED BY CYTOTOXIC T LYMPHOCYTES", IMMUNITY, CELL PRESS, US, vol. 10, no. 6, 1 June 1999 (1999-06-01), pages 673 - 679, XP000890114, ISSN: 1074-7613, DOI: 10.1016/S1074-7613(00)80066-7
PAAL F BRUNSVIG ET AL: "Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer", CANCER IMMUNOLOGY, IMMUNOTHERAPY, SPRINGER, BERLIN, DE, vol. 55, no. 12, 21 February 2006 (2006-02-21), pages 1553 - 1564, XP019422504, ISSN: 1432-0851, DOI: 10.1007/S00262-006-0145-7
Attorney, Agent or Firm:
KOEFOED, Peter et al. (P.O. Box 45Kogle Allé 2, Hørsholm, DK)
Download PDF:
Claims:
CLAIMS

1. A method for treatment of cancer in a human subject in need thereof, said method comprising administering an effective amount of the peptide EARPALLTSRLRFIPK (SEQ ID NO: 1) and an immunological adjuvant to said patient, wherein said patient has received prior radiotherapy and optionally chemotherapy to treat said cancer and where said patient has received the radiotherapy 4-28 days prior to the initiation of treatment with SEQ ID NO: 1.

2. The method according to claim 1, wherein the patient has received both prior radiotherapy and prior chemotherapy. 3. The method according to claim 1 or 2, wherein said cancer non-small cell lung cancer (NSCLC).

4. The method according to any of the preceding claims, wherein SEQ ID NO: 1 is administered intradermally.

5. The method according to claim 4, wherein SEQ ID NO: 1 is administered in the lower abdomen.

6. The method according to claim any one of the preceding claims, wherein said immunological adjuvant is administered at the same site as SEQ ID NO: 1.

7. The method according to any one of the preceding claims, wherein said immunological adjuvant is administered 10-15 minutes prior to SEQ ID NO: 1. 8. The method according to any of the preceding claims, wherein SEQ ID NO: 1 is administered in an amount of 300 nmol.

9. The method according to any one of the preceding claims, wherein said immunological adjuvant is granulocyte macrophage-colony stimulating factor (GM-CSF).

10. The method according to claim 9, wherein GM-CSF is administered in an amount of 75 pg.

11. The method according to any one of the preceding claims, wherein said chemotherapy entails prior weekly administrations of docetaxel 20 mg/m2.

12. The method according to any one of the preceding claims, wherein said radiotherapy entails 3D radiotherapy 2 Gy x 30.

13. The method according to any one of the preceding claims, wherein said patient is subsequently administered the same doses of SEQ ID NO: 1 and immunological adjuvant via the same route according to the following scheme: twice within the week following the first administration, once a week in weeks 2, 3, 4, 6, 8, 10, 14, 18, 22, once at month 6, and once at month 9.

14. SEQ ID NO: 1 for use in a method according to any one of claims 1-13.

15. Use of SEQ ID NO: 1 for use in the preparation of a pharmaceutical composition for the treatment of NSCLC according to the method of any one of claims 1-13.

Description:
TREATMENT OF NON-SMALL CELL LUNG CARCINOMA BY ACTIVE IMMUNOTHERAPY FIELD OF THE INVENTION

The present invention relates to the field of cancer therapy and immunology. In particular, the present invention relates to cancer therapy by active immunotherapy, namely therapy of non-small cell lung cancer by active immunization with a peptide derived from the catalytic subunit of human telomerase.

BACKGROUND OF THE INVENTION

Lung cancer remains the leading cause of cancer death in both men and women worldwide. Non-small cell lung cancer (NSCLC) accounts for about 80% of cases, and most subjects present with inoperable stage III or stage IV disease. Metastatic disease (stage IV) carries a dismal prognosis, with a five year survival of 1%. The successful treatment of stage III patients depends on the control of both local disease and occult metastases. If the disease can be encompassed within an appropriate radiation volume, i.e. stage I-IIIA, curatively intended radiotherapy (>60 Gy) is the treatment of choice. However, the 5-year survival for stage III patients treated with radiotherapy alone is less than 5%. About one third of patients will relapse locally, one third will develop distant metastases and one third will develop both. Several approaches to multimodality treatment have been investigated. These include induction chemotherapy and concurrent chemo-radiotherapy as well as consolidation chemotherapy. However, progress has been limited. Most patients die from relapsed disease, and new treatment strategies are needed.

The development of vaccines for lung cancer has received more attention in recent years. Several large phase III trials are currently underway in NSCLC patients, investigating different vaccine strategies.

The enzyme telomerase is expressed in most human cancers, including NSCLC, and is considered as an attractive target for a universal cancer vaccine. Telomeric DNA confers stability to chromosomes, and normal somatic cells can undergo a limited number of cell divisions because the telomeres are shortened at each mitosis. Tumor cells bypass this biological clock by expressing the enzyme telomerase that synthesises new telomere units.

Peptide GV1001 (SEQ ID NO: 1) consists of 16 amino acids derived from the active site of hTERT. Previously two GV1001 trials have been reported. The GV1001 peptide is recognized on multiple HLA class II molecules encoded by both DP, DQ and DR subloci. This promiscuous HLA-binding profile suggests that the GVlOOl vaccine may be applicable to the general patient population and may elicit a broad T- helper response within each individual. Further, GVlOOl includes nested HLA class I epitopes, facilitating recruitment of CD8+ cytotoxic T cells. In a previous NSCLC vaccine trial, 26 heavily pretreated patients with advanced disease (mostly stage IV) received vaccination with the telomerase peptides 1540 and GVlOOl. No concomitant chemo- or radiotherapy was given. A vaccine-specific T cell response was observed in 13 subjects against GVlOOl and in two patients against 1540, amid no treatment related serious side effects. OBJECT OF THE INVENTION

It is an object of embodiments of the invention to provide a novel treatment regime for patients suffering from NSCLC.

LEGENDS TO THE FIGURE

Fig. 1 : GVlOOl-specific T-cell responses.

PBMCs were obtained prior to start of therapy, at weeks 6, 10, and at every vaccination thereafter. The PBMCs were stimulated once in vitro and tested for proliferation against irradiated PBMCs ± peptide GVlOOl. Columns represent mean SI (response with GV100 divided by response without GVlOOl). If the recorded SI exceeds the upper limit of the respective chart, the exact SI is annotated at the top end of columns. The diagram shows pre- and postvaccination T-cell responses from all evaluable patients. For each subject, the time point with the highest SI is displayed. Responses with SI>2 were considered GVlOOl specific.

Fig. 2: GVlOOl-specific T-cell responses.

PBMCs were obtained prior to start of therapy, at weeks 6, 10, and at every vaccination thereafter. The PBMCs were stimulated once in vitro and tested for proliferation against irradiated PBMCs ± peptide GVlOOl. Columns represent mean cpm or mean SI (response with GV100 divided by response without GVlOOl). If the recorded cpm or SI exceeds the upper limit of the respective chart, the exact cpm or SI is annotated at the top end of columns. B and C show long-term T-cell memory. The diagrams show development of T-cell responses as recorded from follow-up samples. D-F show that samples stored overnight prior to PBMC isolation(*) mostly tested negative, even in subjects where freshly isolated samples tested positive. G depicts parallel testing of PBMC samples isolated upon arrival or stored overnight.

Fig. 3: Survival of patients.

PFS was assessed by Kaplan-Meier analysis, immune responders were compared with non- responders. PFS was defined as clinical endpoint in the protocol described herein because standard treatment after progression was likely to influence OS (stage III patients). The plot shows PFS for study patients with or without a GV1001 T-cell response. Observed PFS is extended for the immune responders.

DISCLOSURE OF THE INVENTION There exists an increasing interest in combining cancer vaccines with conventional therapy. Previous concerns that chemotherapy would preclude immunization are being supplemented with considerations relating to possible synergistic effects.

In many cases, the majority of tumor cells are eliminated by conventional therapy, but the tumor eventually relapses. Cancer vaccines work through different mechanisms and may therefore be effective against cancer cells resistant to chemo- and radiotherapy. Moreover, the chemo-radiation may possibly enhance rather than preclude the immune response. First, tissue damage may induce "danger signals" that provide a pro-inflammatory

microenvironment. Second, there is evidence suggesting that gene products induced by radiation make the tumor more susceptible to a T-cell attack. Third, lung tumors harbor regulatory T-cells considered to inhibit the host immune response, and several studies suggest that chemotherapy may suppress regulatory T-cells and myeloid-derived suppressor cells.

Finally, Docetaxel, which is applied in the present treatment regime (cf. below), may potentially enhance the vaccine response through other mechanisms. The present invention is based on data form a phase II vaccine trial with the telomerase peptide GV1001 (EARPALLTSRLRFIPK, SEQ ID NO: 1) in stage III NSCLC patients. The study evaluated GV1001 vaccination shortly after chemo-radiotherapy.

In particular, results are presented from a phase II trial that investigated vaccination of NSCLC patients with the telomerase peptide GV1001. The clinical study included 23 patients, and no treatment related serious adverse effects were observed. The study demonstrated an 80% immune response rate per protocol. This response rate is considerable compared to most cancer vaccine trials, including those investigating telomerase-based approaches.

Furthermore, the study demonstrated the generation of durable GVlOOl-specific T cell memory responses.

The multimodal approach, the availability of long term data and the association between immune response and clinical outcome are of particular interest.

There are a number of theoretical arguments suggesting that cancer vaccines may be most effective if applied in combinatory drug regimes, but sparse data from clinical studies on how these modalities interact. Interestingly, the frequency of immune responders in the trial underlying the present invention was superior to the present inventors' three ongoing GV1001 trials investigating vaccination as monotherapy. This finding suggests that the applied chemo-radiation did not preclude immunization and may have contributed favourably to the immune response.

While most peptide vaccines represent short HLA-class I matched epitopes, long HLA II- matched peptides like GV1001 may according to the findings herein be particularly suited for combined protocols. Long peptides recruit CD4+ T-helper cells that are known to interact extensively with other immune cells. In irradiated tumor tissue, GVlOOl-specific T-helper cells may engage APCs presenting antigens from apoptotic tumor cells and induce epitope spreading.

The present inventors address this issue in ongoing studies on long term survivors from GV1001 trials and have indeed identified responses against hTERT epitopes outside GV1001. In a new phase I/II trial in NSCLC patients, we plan to combine chemo- or radiotherapy and vaccination with some of these novel hTERT peptides.

There is limited knowledge on the long term development of cancer vaccine responses and on how to design booster vaccine schedules. Some subjects in the presently presented GV1001 trial tested negative in the first post-vaccination immuno-assays, but developed detectable T cell responses after several months of booster injections. Likewise, we have observed in previous studies with GV1001 and other vaccines that T-cell responses appear to be enhanced by booster vaccination. These findings suggest that repeated vaccination over an extended period yields a higher immune response rate and more durable responses. The question of how long to continue booster vaccination, still remains to be clarified.

However, the present inventors have demonstrated that patients immunized for 6-12 months retained GVlOOl-responses in samples obtained up to 43 months after last vaccine. The latter observation suggests that GVlOOl-vaccination may provide durable T cell memory. Regarding this vaccine, it may be sufficient to provide booster injections only to patients without a robust response in follow-up samples.

The cytokine analyses of responses in long-term survivors has shown high levels of key Thl effector cytokines IFNy and TNFa, and low levels of IL-4 and IL-10. This cytokine pattern may suggest a favourable balance between immunity and T cells. If these responses were analysed only for IFNy, IL-4 and/or IL-10, as is common in vaccine trials, they may easily be designated as "Thl". One may therefore note that the present inventors has detected considerable levels of the key Th2 cytokines IL-5 and IL-13. The latter observation is in line with findings in other earlier studies that cytokine profiles in cancer vaccine trials frequently do not follow a Thl/Th2 delineation. It has been suggested that Th2 cytokines may arise in response to powerful immunoactivation. In the long-time survivors observed by the present inventors, the wide range of Thl/Th2 cytokines may also point to a polyfunctional response. Several studies, in particular of infectious diseases, have suggested that polyfunctional cytokine profiles are associated with protective immunity. In previous GV1001 trials without chemotherapy, the frequency of immune responders was similar as assessed by DTH recordings or T-cell assays. By contrast, most subjects in the presently presented trial, where vaccination followed shortly after chemoradiotherapy, were DTH negative. This observation points to a possible immune modulating effect of

chemotherapy on the GV1001 response. Telomerase is expressed by normal stem cells. It is therefore notable that stem-cell related toxicity did not materialize in the trial discussed in the examples below, where GV1001 vaccination was initiated shortly after heavy chemo-radiotherapy. Regarding the long term safety, we have monitored 19 study subjects for more than two years without detecting toxicity (Table 2). Moreover, the long term data from patients in a related study (see below in the end of the present example) has suggested that booster immunization and sustained immune responses exceeding eight years is well tolerated. Continued monitoring of bone marrow samples and peripheral blood counts has not revealed any toxicity. We have also observed retained hematological counts in melanoma and colon cancer patients vaccinated with GV1001 over several years. In all, our data suggest a mild toxicity profile of GV1001 vaccination.

A majority of patients in the NSCLC trial experienced progressive disease in spite of their immune responses. This observation reflects the aggressive nature of advanced NSCLC. On the other hand, we observe durable tumor responses in some subjects and find that nearly all long term survivors belong to the immune responders. We conclude that GVlOOl vaccination immunizes a high proportion of NSCLC patients. The high immunological response rate is encouraging and indicates that the vaccine is applicable to the general patient population without prior HLA typing. Moreover, GVlOOl vaccination induces long term T cell memory against telomerase antigens, while not compromising bone marrow function. The high immune response rate and low toxicity observed in the phase II trial support the concept of combining vaccination with chemo- or radiotherapy. This is of interest for the clinical development of both GVlOOl and other cancer vaccines. Importantly, the present study also provide signs of clinical effect.

A related phase I/II trial in NSCLC patient having advanced disease (performed on patients that have not received concomitant radio- or chemotherapy) has demonstrated a strong association between immune response and survival. We consider that the results warrant further investigation of GVlOOl in NSCLC. The high immune response rate, low toxicity and signs of clinical activity provide a basis for conducting larger randomized trials. It is in this context interesting that 12/13 patients surviving more than 1000 days to date are immune responders. The observed association suggest that a randomized trial evaluating GVlOOl in NSCLC is warranted. In pancreatic cancer, a phase III GVlOOl trial is currently in progress.

Hence based on the above -referenced findings from the phase II clinical trial, the present invention relates to a method for treatment of cancer in a human patient, said method comprising administering an effective amount of the peptide EARPALLTSRLRFIPK (SEQ ID NO: 1) and an immunological adjuvant to said patient, wherein said patient has received radiotherapy and optionally chemotherapy, and wherein said patient has received the radiotherapy 4-28 days prior to said treatment.

As is clear from the present disclosure, this combination therapy has been demonstrated to be promising in NSCLC patients but it is believed that SEQ ID NO: 1 has the potential of inducing a beneficial anti-tumour immunity in patients, who suffer from a number of other cancer types and who are being treated with radiotherapy and/or chemotherapy. As noted herein, it appears that the patients - in spite of the fact that they have been recently treated with aggressive cytostatic/ cytotoxic drugs and/or with radiotherapy - are fully capable of raising immune responses that have a marked effect on the disease progression. Hence, according to the invention, SEQ ID NO: 1 will be useful in combination treatment involving chemotherapy and/or radiotherapy in most if not all cancer types where chemotherapy and/or radiotherapy is/are used to combat the disease. It is preferred that the patients in question have been or are undergoing chemotherapy and that they have received radiotherapy within a period fo 4-28 days prior to initiation of treatment with SEQ ID NO: 1.

In a preferred embodiment, the cancer trated is non-small cell lung cancer (NSCLC). In preferred embodiments of the invention SEQ ID NO: 1 is administered intradermal^, but other convenient administration routes may also be applied. For instance, SEQ ID NO: 1 may be administered subcutaneously, or by other parenteral routes commonly used in the art, such as via the intraperitoneal or intramuscular routes.

A preferred administration location is in the lower abdomen, but the location of

administration may be varied as convenient or practical. This means that the administration may be in the limbs or dorsally.

Typically the immunological adjuvant is administered at the same site as SEQ ID NO: 1. By the "same site" is herein meant both the same location on the human body and the same type of administration. So, if SEQ ID NO: 1 is administered intradermal^ in the lower abdomen, then the adjuvant is also administered intradermal^ in the lower abdomen.

In the practice of the invention said immunological adjuvant is typically administered shortly prior to administration of SEQ ID NO: 1, such as between 1-30 or 2-29 or 3-28 or 4-27 or 5- 26 or 6-25 or 7-26 or 8-25 or 9-24 minutes prior to SEQ ID NO: 1. Preferably the adjuvant is administered 10-15 minutes prior to SEQ ID NO: 1. The amount of SEQ ID NO: 1 administered must be effective, which means that the amount is at leat 10 nmol per administration, such as at least 50 nmol, or at least 200 or at least 250 nmol. On the other hand, the amount administered is often at most 2000 nmol, such as at most 1500 nmol, at most 1000 nmol, or at most 800 nmol, such as at most 500 nmol. In especially preferred embodiments, the an amount is about 300 nmol. The adjuvant is any convenient immunological adjuvant, but especially immunostimulating adjuvants are relevant. Examples of suitable immunological adjuvants are discussed extensively in WO 98/20027. A particularly interesting immunologically adjuvant is granulocyte macrophage-colony stimulating factor (GM-CSF) - this adjuvant is in certain embodiments of the invention administered in an amount of 75 pg per administration, but may be administered in both higher or lower dosages as determined by testing the potency of GM-CSF when administered via a particular route and in a particular location on the body. As discussed above, the patients treated according to the present invention have been previously treated with chemotherapy and radiotherapy. Any chemotherapeutical regime useful in treatment of NSCLC may have been used, but in the practice of the current invention, the beneficial effects have been observed when immunizing with SEQ ID NO: 1 after treatment with docetaxel, in particular after weekly administrations of docetaxel 20 mg/m 2 (where the m 2 indication is skin surface area of the treated individual).

Also the radiotherapeutic treatment regime may be any regime convenient in the treatment of NSCLC, but in the practice of the present invention, the results reported below was obtained after radiotherapy that consisted of 3D radiotherapy 2 Gy x 30. The method of the invention typically entails repeated immunizations after immunization 1 in order to induce an effective immune response. The immunization scheme followed in the examples below entails that patients are subsequently administered the same doses of SEQ ID NO: 1 and immunological adjuvant via the same route as in the first immunization and according to the following scheme: twice within the week following the first administration, once a week in weeks 2, 3, 4, 6, 8, 10, 14, 18, 22, once at month 6, and once at month 9. However, such immunization schemes may be optimized, and as indicated in the examples, there is a possibility that the number of follow-up or booster immunizations can be reduced - a skilled immunologist will be capable of determining from further experiments an optimized immunization scheme. Alternatively, the immunological status of the individual patients may be monitored and based on result from this monitoring the immunization protocol for each patient may be adjusted individually.

The invention also relates to GV1001 (SEQ ID NO : 1) for use in the method disclosed herein and as set forth in the claims. Further the invention also relates to use of SEQ ID NO: 1 for the preparation of a pharmaceutical composition for the treatment of NSCLC according to the method of the invention disclosed above and as defined in the claims.

Definitions

"GV1001" denotes the peptide EARPALLTSRLRFIPK (SEQ ID NO: 2), which is derived from the amino acid sequence of human telomerase protein (hTERT).

"Radiotherapy" generally denotes tumour treatment by use of ionizing radiation, which can be applied locally or systemically. The exact type of radiation therapy useful differs from cancer type to cancer type and is generally well known to the skilled person. "Chemotherapy" is used generally for treatment of cancer patients with a variety of cytotoxic or cytostatic drugs. Thus in the present disclosure, chemotherapy for instance denotes anticancer therapy using the following groups of drugs:

Anti-metabolites (L01B) mimic purines (e.g. azathioprine and mercaptopurine) or pyrimidines, which are the building-blocks of DNA. The anti-metabolites prevent these substances from becoming incorporated into DNA during the "S" phase of the cell cycle, stopping normal development and division. They also affect RNA synthesis.

Plant alkaloids and terpenoids (L01C). These alkaloids prevent microtubule function. The main examples are vinca alkaloids and taxanes. Vinca alkaloids (L01CA) are derived from the Madagascar periwinkle, Catharanthus roseus (formerly known as Vinca rosea) and include vincristine, vinblastine, vinorelbine, and vindesine.

Podophyllotoxin (L01CB) is a plant-derived compound that is said to help with digestion as well as used to produce two other cytostatic drugs, etoposide and teniposide. They prevent the cell from entering the Gl phase (the start of DNA replication) and the replication of DNA (the S phase).

Taxanes (L01CD) are derived from the natural product paclitaxel, originally known as Taxol and first derived from the bark of the Pacific Yew tree. Docetaxel is a semisynthetic analogue of paclitaxel. Taxanes enhance stability of microtubules, preventing the separation of chromosomes during anaphase of the cell cyclus.

Topoisomerase inhibitors (L01CB and L01XX). These include type I topoisomerase inhibitors such as the camptothecins irinotecan and topotecan, and the type II inhibitors such as amsacrine, etoposide, etoposide phosphate, and teniposide.

Cytotoxic antibiotics (L01D) include actinomycin (L01DA01), anthracyclines such as doxorubicin (L01DB01), daunorubicin (L01DB02), valrubicin, idarubicin, epirubicin (L01DB03), and other cytotoxic antibiotics such as bleomycin (LOIDCOI), plicamycin (L01DC02), and mitomycin (L01DC03).

An "immunological adjuvant" has its usual meaning in the art: A substance or composition which, when administered to an individual assists in the induction/elicitation of a specific immune response towards an antigen. A preferred adjuvant in the present invention is GM- CSF. EXAMPLE

MATERIALS AND METHODS Patients and study protocol

The primary objective of the phase II trial was immunological response. Toxicity and time to progression were secondary objectives. Twenty-three subjects with inoperable stage IIIA/B NSCLC were enrolled between November 2006 and July 2008 from 3 different centres in Norway. Twelve patients were enrolled at The Norwegian Radium Hospital, four at St Olav's Hospital and seven at The Southern Hospital of Norway, Kristiansand. The trial was approved by the Norwegian Medicines Agency, the Regional Committee for Medical Research Ethics and the Hospital Review Board. It was performed in compliance with the World Medical

Association Declaration of Helsinki. Signed informed consent was obtained from all patients.

The study population had been treated with weekly docetaxel 20mg/m2 and 3 D radiotherapy 2 Gy x 30 within the last 4 weeks. Subjects with metastatic disease were excluded based on a pre-study CT scan of the thorax and upper abdomen and an MRI scan of the brain. The eligibility criteria also included Eastern Oncology Group (ECOG) performance status 0-2, age > 18 years, WBC > 1.5 x 10 9 /L; platelets > 100 x 10 9 /L, Hb > 9g/dL (> 5.6 mmol/L);

creatinine≤ 140 pmol/l (1.6 mg/dl), bilirubin≤ 20% above the upper limit of normal, ASAT and ALAT≤ 1.5, the upper limit of normal albumin > 2.5 g/L. Exclusion criteria included a history of other prior malignancy, with the exception of curatively treated basal cell or squamous cell carcinoma of the skin or cervical cancer stage IB, active infection requiring antibiotic therapy or significant cardiac or other medical illness, such as severe congestive heart failure, unstable angina, serious cardiac arrhythmia, serious adverse reactions to vaccines such as anaphylaxis. Patients with known autoimmune diseases or who test positive for hepatitis B, C or HIV were excluded from the study. Study design

The strategy behind the study design was to pave the way for a phase III trial in stage III NSCLC patients, evaluating the vaccine within a multimodal treatment regime. The dosage of GV1001 was based on data from our previous dose-escalation trials in NSCLC and pancreatic cancer. The chemoradiotherapy represented institutional standard treatment in 2006 for inoperable stage III NSCLC. Our decision to include 20 evaluable patients was based on the main study objectives: to show that combined treatment with chemoradiotherapy and GVlOOl is feasible and may yield immunization, to provide safety data and to obtain an estimate for PFS and immune response rate. In a given sample size, the number of subjects with immune response and serious adverse events (SAE) will follow a binomial distribution. Statistical calculations based on n=20 and a binomial distribution shown. The probability of detecting 5 or more immune responders was 99.8%, assuming a true response rate of 54% as observed in the phase I/II study. The probablility of detecting 1 or more SAE was 87.8%, assuming a true SAE frequency of 10%.

Treatment

In the trial, vaccination with GV 1001 started within 4 days to 4 weeks following the last radiotherapy treatment. Immunization was given in week 1 (Monday, Wednesday and Friday) and once in each of weeks 2, 3, 4, 6 8 and 10. A boost immunization was given in each of weeks 14, 18, 22, at 6 month and at 9 months. GVlOOl (300 nmol peptide in 0.20 ml saline) was injected intradermally (i.d.) in the lower abdomen. GM-CSF (75 pg Leukine; Bayer, Oslo, Norway) was injected at the same site 10-15 minutes prior to GVlOOl. Peptides

The vaccine peptide GVlOOl corresponds to the 16 amino acid residue 611-626

(EARPALLTSRLRFIPK; SEQ ID NO: 1) of hTERT. GVlOOl was supplied by Pharmexa

(Horsholm, Denmark). Manufacturing was in compliance with GMP. RAS-peptide 508 (KRAS 52-70, Q61H; Norsk Hydro, Norway) served as a negative control in T-cell assays. T cell cultures and assays

Peripheral blood mononuclear calls (PBMCs) were obtained prior to start of therapy, at weeks 6, 10 and at every vaccination thereafter. The PBMCs were isolated and frozen as previously described. Thawed PBMCs were stimulated once in vitro with the vaccine peptide prior to T cell assays, as described earlier . At this initial stimulation, the PBMCs were cultured with GVlOOl (25 pmol/l) for 7 ot 10 days, with addition of IL-2 (10 U/ml) from day 3.

T-cell proliferation assays ( 3 H Thymidine) were performed essentially as previously described. Pre- and post-vaccination samples were analyzed in parallel for response to peptide stimulation. Irradiated autologous PBMCs were used as antigen presenting cells (APCs).

Stimulation with Staphylococcal enterotoxin C (SEC) was used as positive control and as a measure of immunocompetence. All patients responded to SEC. T cell cultures were tested in triplicates. SEM was usually below 10%. Proliferation counts after stimulation with the irrelevant peptide (K-RAS 508) were generally not significantly different from controls without peptide. T cell responses were considered antigen-specific when the stimulatory index (SI; response with antigen divided by response without antigen) was above 2. Bioplex cytokine analyses were done on supernatants harvested 48 hours after T-cell stimulation, according to the manufacturer's protocol (Bio-Rad Laboratories). Supernatants were analysed in duplicates/triplicates, each parallel kept separate through T-cell stimulation and Bioplex assays.

Delayed-Type Hypersensitivity Delayed-type hypersensitivity (DTH) skin test was performed at baseline, at weeks 2, 3, 4, 6, 10 and at the time of later vaccinations. For DTH testing, 60 nmol GV1001 in 0.10 ml saline was injected i.d. at a site separate from the site of vaccination, without GM-CSF. The patients registered the DTH skin reaction 48 hours after administration. A positive DTH test was defined as an erythema/induration with average diameter > 5 mm. Clinical evaluation

Adverse drug reactions and ECOG performance status were assessed at each visit. Blood screening and a general physical examination were performed at start of vaccination (week 1), weeks 2, 3, 4, 6, 8, 10 and all later vaccinations. CT scans were performed before start of vaccination, at week 14 and every third month thereafter. Progression-free-survival was defined as main clinical end point in the CTN-2006 protocol, because overall survival was likely be influenced by standard treatment after progression. Radiation fibrosis is in general difficult to reliably distinguish from tumor. At start of vaccination, the patients had residual CT lesions after chemo-radiotherapy that may, or may not, include viable tumor tissue. The terms complete response, partial response and stable disease were therefore not applicable. Progressive disease was defined as new or progressing lesions, identified by CT scans, bronchoscopy and/or biopsy. Statistics

PFS was calculated from start of vaccination. Kaplan-Meyer/log-rank analysis was applied from comparing immune responders versus nonimmune responders with regard to PFS. To assess whether the immune response represented an independent prognostic factor, Cox regression with enter analysis was conducted. Disease stage represented the most important identifiable prognostic factor, apart from immune response. The subjects were staged as stage IIIA or IIIB.

RESULTS

Patient characteristics and adherence to treatment Patient characteristics and treatment details are listed in Table 1. At the end of 2007, Bayer withdrew liquid GM-CSF (Leukine) from the markets. This led to a sudden shortage until the lyophilized product was delivered. Two patients at Radiumhospitalet therefore received one of their GV1001 injections without immunological adjuvant GM-CSF. At St Olav's hospital, two other patients missed the combined vaccination twice each (week 8 and 10). The study monitoring panel decided that patients missing GM-CSF at two vaccinations (No. 203 and No. 204) were to be replaced and excluded from the per protocol analysis. Patient No. 110 was also not evaluable per protocol, because she was withdrawn at week 8 due to an abscess in the right lung and progression of disease.

Safety The safety population includes all patients who received at least one vaccination (n = 23). A total of 323 vaccine doses were administered (8-21 doses per patient). Seven serious adverse events were reported, from six patients. All seven events were regarded as related to underlying disease, not to study therapy. One event, a bronchiolar fistula, was initially reported as probably related to a study drug. However, a bronchoscopy with biopsy demonstrated that the fistula was due to tumor relapse. Immune response

A GV1001- specific T cell response was demonstrated in 16 patients after vaccination, compared to no patients in pre-vaccination samples (Fig. 1). A positive DTH response was observed in one patient only. Three subjects were not evaluable according to the protocol (see above). The immunological response rate was 70% by intention to treat (ITT) analysis, and 80% per protocol.

To achieve a sustainable clinical effect, development of T cell memory is likely to be required. We therefore provided booster vaccines and monitored the long term development of immune responses. Follow-up samples where obtained in 15/16 immune responders. The results demonstrated a durable GVlOOl-specific T cell response in 13/15 subjects, with a maximum observation period of 91 weeks (Table 2 and Figs. 2A-B).

Comparing the different centres, we recorded a GVlOOl-specific T-cell response in 9/12 patients from Radiumhospitalet Oslo (9/11 per protocol), 1/4 patients (1/2 per protocol) from St Olav's Hospital Trondheim and 6/7 from Kristiansand. The samples from Trondheim and Kristiansand were sent to Oslo for T cell analyses. Some of the first samples were stored overnight prior to PBMC isolation. Samples stored overnight gave only negative results. We therefore decided to isolate subsequent samples from the same patients immediately upon arrival. Interestingly, most previously negative patients then tested positive. Moreover, later samples from some patients were stored overnight and again tested negative (Figs. 2C-E). We also tested samples stored the same day or overnight in parallel and observed distinctly stronger responses in freshly stored samples (Fig. 2F). These observations illustrate the complexity of managing a multi-center trial with T cell analyses, and suggest that T cell data may be misleading if analyses are performed on samples not optimally handled.

Clinical response Table 2 lists progression-free survival (PFS), overall survival and site of relapse. PFS was the clinical end point per protocol and was assessed by CT scans at three month intervals. To date, tumor progression has been recorded for 17/23 patients in the ITT population (median PFS 357 days). Five out of six patients without evidence of relapse are immune responders. Considering all included patients, immune responders recorded increased PFS compared to non-responders, with a median of 371 days versus 182 days (Fig. 3).