Download VCG1 : preliminary results

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Immunocontraception wikipedia , lookup

Immunomics wikipedia , lookup

Management of multiple sclerosis wikipedia , lookup

Autoimmune encephalitis wikipedia , lookup

Vaccination policy wikipedia , lookup

Multiple sclerosis research wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Multiple sclerosis signs and symptoms wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Vaccination wikipedia , lookup

Transcript
Clavreul et al.
Autologous tumor cell vaccination plus infusion of GM-CSF by a programmable
pump in the treatment of recurrent malignant gliomas
Anne Clavreula, Nicole Piardb, Jean-Yves Tanguyc, Eric Gamelind, Marie-Christine
Rousselete, Pierre Leyniad, Philippe Meneia
a
Département de Neurochirurgie, CHU, Angers, F-49933 France; INSERM U646,
Angers, F-49100 France; UNAM, Angers, F-49100 France
b
c
Département de Radiologie, CHU, Angers, F-49933 France
d
e
EFS, Angers, F-49100 France
Centre Paul Papin, CRLCC, Angers, F-49933 France
Laboratoire Pathologie Cellulaire et Tissulaire, CHU, Angers, F-49933 France
Corresponding author:
Dr Anne Clavreul
Département de Neurochirurgie
CHU
49933 Angers, France
Tel: +33 241 354822
Fax: +33 241 354508
E-mail: [email protected]
This work was supported by a PHRC from National Health Department and a grant from
the Ligue Départementale de Lutte contre le Cancer.
1
Clavreul et al.
Abstract
This phase I study reports on the safety and feasibility of autologous cell vaccination
combined with infusion of GM-CSF by a programmable pump in the treatment of
recurrent malignant gliomas. The trial enrolled nine patients who had previous surgery,
radiation and were successfully weaned off steroids. Unfortunately, only five patients
completed the protocol and were monitored for side effects, local reactions, delayedtype hypersensitivity (DTH) responses and survival. The treatment was well tolerated.
Two patients developed DTH reactions after vaccination and three patients had an
unusually long survival without any other treatment. Despite the small number of
patients treated, this study is informative and encouraging. The programmable pump is a
promising tool to infuse cytokines subcutaneously for vaccination purposes.
Nevertheless, this study as others highlights also the specific difficulties encountered in
such vaccination paradigms for the treatment of glioma.
Key Words: GM-CSF, immunotherapy, malignant glioma, vaccination
2
Clavreul et al.
1. Introduction
Malignant gliomas have a poor prognosis despite aggressive treatment using surgery,
radiotherapy and chemotherapy 1, 2. One therapy emerging over the last few years is
active immunotherapy to initiate T-cell-mediated antitumor immune responses 3, 4. This
therapy offers several advantages such as tumor specificity with low side effects and a
durable antitumor effect owing to the phenomenon of immunologic memory. Two main
active immunotherapy strategies are used for glioma patients: peripheral injections of
irradiated autologous tumor cells (ATC) along with an adjuvant for immune stimulation
or vaccinations with autologous dendritic cells primed with tumor antigens ex vivo 3, 4.
The use of irradiated ATC vaccines is considered an advantage, given the lack of
knowledge regarding the relevant tumor associated antigens (TAAs) in glioma.
Furthermore, injection of these irradiated ATC allows an antigenic treatment in vivo
without manipulation of dendritic cells ex vivo. To render these ATC more
immunogenic, different cytokines and chemokines have been used as immune
adjuvants. One of the most frequently used cytokine is granulocyte-monocyte colonystimulating factor (GM-CSF) which is a potent activator of dendritic-cell antigen
presentation, and participates in the initiation of danger signals needed to activate the
immune system, break tolerance, and develop an antitumor immune response 5. Several
preliminary studies using ATC plus GM-CSF alone showed promising results in brain
tumors 6-12. To date, in clinical studies, administration of GM-CSF to the vaccination
site is achieved principally by ATC, allogeneic tumor cells or even normal bystander
cells, genetically modified to express GM-CSF 13-17. This approach allows a prolonged
GM-SF delivery but is time and labor consuming 14. Furthermore, the safety of this
method still needs to be addressed. Another GM-CSF delivery approach consists in the
3
Clavreul et al.
use of pump which allows a continuous infusion of the cytokine with a good safety
profile 9, 18, 19.
This clinical trial was therefore designed to investigate the safety and feasibility of
vaccination with ATC and infusion of GM-CSF by a programmable pump in the
treatment of recurrent malignant gliomas.
2. Materials and Methods
2.1. Patient eligibility
This trial was an open, nonrandomized phase-I study. The protocol was approved by the
Committee on the Right of Human Subjects and the biotherapy department of French
Health Products Safety Agency. All patients provided informed consent before
treatment. Inclusion criteria were patients presenting a recurrent grade III or IV
malignant glioma that was amenable to surgical resection, an age between 18 and 68
years, a Karnofsky index (KI) > 60. Patients must have been previously treated by
conventional radiotherapy (60 Gy) and tapered off steroids and cytotoxic drugs for at
least one month at the time of vaccination. Exclusion criteria included pregnancy, severe
pulmonary, cardiac or other systemic diseases.
2.2. Assessment of extent of tumor resection before vaccination
In the 72 h following the surgery, a MRI was performed including T1 with and without
gadolinium, T2 and Fluid Attenuated Inversion Recovery (FLAIR) weighted sequences.
The extent of resection was defined as biopsy (less than 10% resected), sub-total
resection (10% to 90% resected) and gross-total resection (more than 90% resected).
The resection volume has been calculated on MRI slices.
2.3. Autologous tumor culture
4
Clavreul et al.
After surgery, tumor tissue was mechanically disaggregated into a suspension and then
passed through nylon meshes of decreasing pore size. The cell suspension was cultured
in Dulbecco’s modified Eagle’s medium (DMEM) (Lonza, Verviers, Belgium)
supplemented with 10% human AB serum or FCS (Hyclone, PerbioScience, Bredières,
France) and 1% penicillin/streptomycin (Lonza). Following in vitro expansion, cells
were irradiated at 45 Gy using a 137Cs source (EFS, Angers, France), checked for
viability by trypan blue exclusion and stored at -80°C. Prior to inoculation, culture
media were tested for bacterial contamination.
2.4. Tumor vaccine preparation
On the day of vaccination, the cells were thawed, washed two times with physiological
serum and counted. Between 2 to 5106 ATC in 600 µl of physiologic serum were
injected subcutaneously.
2.5. GM-CSF infusion
RhGM-CSF (Leucomax®, Novartis, Schering-Plough, Levallois Perret, France or
Leukine®, Berlex, Seattle, United States) was infused at the site of cell inoculation. The
infusion was performed using a programmable pump MiniMed 407C and standard
insulin infusion sets, Sof-Set (Medtronic MiniMed, Northridge, United States).
Reservoirs containing GM-CSF were changed weekly and the remaining GM-CSF was
quantified by ELISA (R&D Systems, Lille, France).
2.6. Vaccination shedule
Two sets of vaccination protocols have been performed (Figure 1). Protocol 1 consisted
in four subcutaneous vaccinations with ATC every 7 days in abdomen and a continuous
infusion of GM-CSF (10 µg/24 h) during 28 days. Protocol 2 was composed of four
subcutaneous vaccinations with ATC every 21 days close to cervical lymph nodes and
5
Clavreul et al.
an infusion of GM-CSF (20 µg/24 h) 3 days before and during 14 days after each
vaccination.
2.7. Evaluation of clinical status
Patients were followed by clinical and MRI examination and blood cell counting (BCC).
Toxicity was monitored using the National Cancer Institute Common Toxicity Criteria.
Response to the treatment was analyzed using Mac Donald criteria 20: complete response
(total disappearance of all enhancing tumor, patient stable or improved), partial response
(50% or greater reduction in size, patient stable or improved), stable (reduction of 050% in tumor size, patient stable or improved), progressive disease (25% or greater
increase in size or any new tumor, or patient worse).
2.8. Delayed-type hypersensitivity reaction
Delayed type hypersensitivity (DTH) was tested before and after the final vaccination.
For this, an intradermal ATC inoculation (1106 cells) was performed in the shoulder,
followed by a cutaneous punch biopsy 48 h later. Formalin-fixed, paraffin-embedded
sections of the cutaneous samples were stained with hematoxylin-phloxin-saffron and
studied by immunohistochemistry for astrocyte-, T-, B-, monocyte-macrophage-,
Langerhans cell-, and NK cell-differentiation antigens using a streptavidin-biotin
peroxidase method on a DAKO automatic immunostaining device. The specificity and
source of the antibodies are listed in Table 1.
2.9. Autoimmunity detection
Sera of patients were collected before and after the treatment to determine if
autoimmunity develops against myelin. Autoantibodies directed against myelin
associated glycoprotein (MAG) were quantified by ELISA according to the
manufacturer’s instructions (Bühlmann, Mulhouse, France).
6
Clavreul et al.
2.10. Statistical analysis
The prevaccination and postvaccination data were compared using the Student’s t-test
modified for small samples. Statistical significance was determined at P < 0.05 level.
3. Results
3.1. Patient characteristics
Nine patients were enrolled in this phase I trial between 2001 and 2005, seven in
Protocol 1 and two in Protocol 2. Their characteristics are summarized in Table 2. They
were four women and five men with an age range from 33 to 65 years (mean 49.9
years). All patients have been previously treated by radiotherapy, six by chemotherapy.
Histopathological diagnosis made after second surgery was the same as the one made
after the initial surgery: glioblastoma (n=6), anaplastic oligodendroglioma (n=2) and
anaplastic oligoastrocytoma (n=1). The extent of resection was a sub-total resection for
six patients and a gross-total resection for three patients (Table 2).
3.2. Vaccine preparation
Preparation of irradiated tumor cells was initiated after a mean interval of 4.5 weeks
(1.4 to 9 weeks) after surgery and the cell viability was consistently 80-90% before
freezing. For 6/9 patients, cell yields were not sufficient to allow four vaccinations.
3.3. Vaccine administration
Among the nine patients enrolled in this trial, four have not been vaccinated. Three
patients (n°3, n°6 and n°9) because tumors have progressed and clinical status worsened
during the time of the tumor cell culture and one patient (n°7) because the Health
Agency has suspended the utilization of the human serum produced by our first
supplier, a biotechnology company. Among the four patients treated in Protocol 1, three
7
Clavreul et al.
received two vaccinations (patients n°1, n°4 and n°5) and one three vaccinations
(patient n°2) (Table 2). The patient n°8 treated in Protocol 2 received four vaccinations.
Between 2 to 5106 ATC (mean 3.0106  1.1106) were inoculated for vaccination.
3.4. GM-CSF infusion
A dose of 10 µg/24 h (Protocol 1) or 20 µg/24 h (Protocol 2) was administrated to
vaccinated patients. The infusion device was well tolerated. GM-CSF in the pump
reservoir was changed every week and the stability of the cytokine was analyzed by
ELISA. For patients receiving a dose of 10 µg/24 h, the initial concentration of GMCSF in the reservoir was 30 µg/ml and after 7 days, the remaining concentration of GMCSF was 26.5  3.2 µg/ml.
3.5. Safety
The two serious adverse effects reported were due to the surgery, one hemiplegia of
vascular origin (patient n°4) and one post operative hematoma necessitating a
reintervention (patient n°7). The others side effects observed during the treatment were
minor and can be attributed to GM-CSF: vomiting (n=1), asthenia (n=1), astheniaanorexia-arthralgia (n=1), episode of shivering, cyanosis, low blood pressure, vomiting,
fever, 15 min after the first DTH (n=1). An increase of peripheral blood eosinophils was
observed in the four patients treated in Protocol 1 (1.18%  0.12% before vaccination to
6.53%  1.64% three weeks after the first vaccination; P < 0.05). This increase was also
noticed in the patient treated in Protocol 2 but in a less important manner. No cutaneous
complication was observed at the inoculation site. There was no evidence of growth of
tumor cells at the vaccine site in any patient. No sign of leucoencephalopathy (clinical
or on the MRI-FLAIR sequence) was detected, nor autoantibodies against MAG.
3.6. Response to immunization
8
Clavreul et al.
Between 0.6 to 1.6106 ATC (mean 1.1106  0.4106) were inoculated for DTH before
and after the vaccination schedule. No patients developed local DTH reactions before
vaccination. Punch biopsies showed a normal epidermis and, in the superficial dermis, a
mild perivascular accumulation of T lymphocytes, mostly CD4 positive, with rare
Langerhans cells and no or exceptional B lymphocytes. In the reticular dermis, rare
neutrophils were observed close to a few glial cells in patients n°1 and n°2. In patients
n°4, n°5 and n°8, some glial cells and GFAP positive fragments were embedded in a
larger inflammatory reaction composed of macrophages and polymorphonuclear
leukocytes (mostly neutrophils and a few eosinophils) with some leukocytoclasis. There
were no NK cells. After vaccination, the punch biopsy of two patients (n°5 and n°8)
showed an increase of perivascular T cells and Langerhans cells in the superficial
dermis compatible with a DTH reaction (Figure 2). Other patients did not develop
cellular infiltrates characteristic of DTH despite the fact that a small induration (5 mm)
with erythema developed to the site of the inoculation for patient n°4.
3.7. Clinical response
Clinical responses are detailed in Table 2 and in Figure 3. Three patients (n°4, n°5 and
n°8) were clinically stable after vaccination. They presented an unusually long survival,
respectively 62, 42 and 88 weeks after the second surgery without corticoids and
adjuvant treatments. The two other patients (n°1 and n°2) had a progressive disease
after vaccination.
4. Discussion
This phase I study was designed to investigate the safety and feasibility of vaccination
using irradiated ATC and infusion of GM-CSF in patients with recurrent malignant
9
Clavreul et al.
glioma. While the concept of this trial is quite similar in many ways to previous studies,
the novel aspect of this study was the use of programmable pump with standard insulin
infusion sets to deliver GM-CSF at the vaccination inoculation site. This infusion
technology allows a sustained subcutaneous delivery in a flow as small as 1 µl/h. The
principal concern with this technology was the instability of GM-CSF and the
possibility of its degradation in the reservoir at room temperature. For this reason,
reservoirs were changed every week. Interestingly, in this study, after 7 days at room
temperature, GM-CSF showed a satisfactory stability. Furthermore, GM-CSF
administration induced an increase of peripheral blood eosinophils. This hyper
eosinophilia has been already described after subcutaneous administration of low doses
of GM-CSF (as 7.5 to 45 µg/day for 10 days) 21. These results indicate that
programmable pump could ensure the release of a biologically active GM-CSF during
vaccination avoiding the use of ATC retrovirally transduced with GM-CSF which is
time and labor consuming 14.
This clinical trial was originally designed to enroll fifteen patients. However, we closed
the trial after nine patients due to the unavailability of GM-CSF and the difficulty to
enclose patients tapered off steroids. Unfortunately, among the nine patients enrolled,
only five patients completed the protocol because in others patients, tumors had
progressed and clinical status worsened during the time of the tumor cell culture. This
small number of patients does not allow drawing conclusions about the safety and
clinical response of vaccination with ATC and infusion of GM-CSF by pump.
Furthermore, we could not comment meaningfully on the efficacy between the Protocol
1 and 2. Nevertheless, three treated patients (patients n°4, n°5 and n°8) had an unusually
long survival without any other treatment (respectively 61, 42 and 88 weeks after the
10
Clavreul et al.
second surgery). Mean survival in glioblastoma after recurrence is 23 weeks after
surgery alone and 31 weeks after surgery plus local chemotherapy 22. Interestingly, these
three patients developed a larger inflammatory reaction at their first DTH site and had a
moderate size of the recurrence at the time of vaccination. Furthermore, two of these
three patients (n°5 and n°8) showed a histological DTH reaction after vaccination
suggesting the development of a peripheral antitumor immune response. The two other
treated patients (n°1 and n°2) did not show prolong survival nor the development of an
immune reaction after vaccination. However, it is important to note that patient n°1 was
vaccinated despite a low KI and an important tumor recurrence. Patient n°2 was
vaccinated although its MRI showed, at the time of the first vaccination, a recurrence at
distance of the surgical cavity.
If these results are encouraging, they point out several issues linked to the methodology
of vaccination against glioma. Firstly, the difficulty of re-intervention, especially when
the goal is to perform a sub-total resection. Two reported serious adverse effects were
due to this second surgery. Unfortunately, the resection is necessary since extent of
disease at enrollment seems to correlate strongly with poor response to active
immunotherapy 14, 23 . Secondly, this study, as others, shows the difficulty to rapidly and
reliably obtain sufficient ATC for vaccination 14, 24, 25. In our study, a mean interval of
4.5 weeks was necessary to prepare irradiated tumor cells after surgery. The use of
cultured ATC as vaccines seemed to us a good compromise since no glioma-specific,
immunologic-relevant TAAs have been identified. While there is a lot of evidence in the
literature that tissue culture conditions change the phenotype of tumors, we showed in a
previous study that our culture conditions preserved at early passage cultures the cell
population of interest present in the original tumor and TAA expression 26. However, it
11
Clavreul et al.
is clear that efforts should be made to obtain sufficient cell number in a relatively short
period of time to avoid large recurrence. To increase the cell yield, some clinical studies
used frozen tumor digest or formalin-fixed tumor tissue for vaccination with no cultured
cells 6, 27, 28. However, these preparations contain tumor cells but also normal cells
which could induce autoimmunity after their injection. Besides, the induction of lethal
experimental allergic encephalomyelitis has been described in primates and guinea pigs
after vaccination with human glioblastoma tissue 29. In our study, we do not have
noticed demyelination with MRI-FLAIR sequence, which is considered the most
sensible sequence to show demyelination within the white matter of the cerebral
hemispheres 30, nor autoantibodies directed against MAG in sera of patients. Another
alternative to solve the problem of time-consuming ATC expansion is the use of
synthetic glioma peptides or allogeneic tumor cells as vaccines 31-34. Finally, despite the
high number of active immunotherapy clinical trials conducted in malignant gliomas,
there is still a lack of definite proof for efficacy 3. In fact, this clinical trial as others
shows that induction of a peripheral antitumor immune response is possible in glioma
patients but not sufficient to preclude disease progression. Several studies point out the
importance of the immunosuppressive tumor microenvironment in the glioma
immunotherapy resistance 3, 19, 35. Counteracting this immunosuppressive tumor
microenvironment is essential in the success of the future anti-glioma vaccines.
Acknowledgments: We are grateful to Medtronic Minimed, Northridge, United States,
for the kind gift of pumps and infusion sets. We thank the Laboratory of Cellular
Biology (Prof A. Barthelaix/L. Denéchaud), the Laboratory of Bacteriology (Prof F.
Lunel-Fabiani) and the Laboratory of Hematology (Prof M. Zandecki/Dr F. Genevieve),
12
Clavreul et al.
CHU, Angers, France, for technical assistance. We also thank the members of the
Clinical Trial Department, Cancer Center Paul Papin, Angers, France, for the facilities
provided.
13
Clavreul et al.
References
1. Westphal M, Hilt DC, Bortey E, et al. A phase 3 trial of local chemotherapy with
biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary
malignant glioma. Neuro Oncol 2003;5:79-88.
2. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and
adjuvant temozolomide for glioblastoma. N Engl J Med 2005;352:987-96.
3. Okada H, Kohanbash G, Zhu X, et al. Immunotherapeutic approaches for glioma.
Crit Rev Immunol 2009;29:1-42.
4. Yamanaka R. Dendritic-cell- and peptide-based vaccination strategies for glioma.
Neurosurg Rev 2009;32:265-73; discussion 73.
5. Chang DZ, Lomazow W, Joy Somberg C, et al. Granulocyte-macrophage colony
stimulating factor: an adjuvant for cancer vaccines. Hematology 2004;9:207-15.
6. Plautz GE, Barnett GH, Miller DW, et al. Systemic T cell adoptive immunotherapy of
malignant gliomas. J Neurosurg 1998;89:42-51.
7. Plautz GE, Miller DW, Barnett GH, et al. T cell adoptive immunotherapy of newly
diagnosed gliomas. Clin Cancer Res 2000;6:2209-18.
8. Lefranc F, Cool V, Velu T, et al. Granulocyte macrophage-colony stimulating factor
gene transfer to induce a protective anti-tumoral immune response against the 9L rat
gliosarcoma model. Int J Oncol 2002;20:1077-85.
9. Wallenfriedman MA, Conrad JA, DelaBarre L, et al. Effects of continuous localized
infusion of granulocyte-macrophage colony-stimulating factor and inoculations of
irradiated glioma cells on tumor regression. J Neurosurg 1999;90:1064-71.
14
Clavreul et al.
10. Yu JS, Burwick JA, Dranoff G, et al. Gene therapy for metastatic brain tumors by
vaccination with granulocyte-macrophage colony-stimulating factor-transduced tumor
cells. Hum Gene Ther 1997;8:1065-72.
11. Herrlinger U, Kramm CM, Johnston KM, et al. Vaccination for experimental
gliomas using GM-CSF-transduced glioma cells. Cancer Gene Ther 1997;4:345-52.
12. Sloan AE, Dansey R, Zamorano L, et al. Adoptive immunotherapy in patients with
recurrent malignant glioma: preliminary results of using autologous whole-tumor
vaccine plus granulocyte-macrophage colony-stimulating factor and adoptive transfer of
anti-CD3-activated lymphocytes. Neurosurg Focus 2000;9:e9.
13. Ward JE, McNeel DG. GVAX: an allogeneic, whole-cell, GM-CSF-secreting
cellular immunotherapy for the treatment of prostate cancer. Expert Opin Biol Ther
2007;7:1893-902.
14. Parney IF, Chang LJ, Farr-Jones MA, et al. Technical hurdles in a pilot clinical trial
of combined B7-2 and GM-CSF immunogene therapy for glioblastomas and
melanomas. J Neurooncol 2006.
15. Soiffer R, Hodi FS, Haluska F, et al. Vaccination with irradiated, autologous
melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating
factor by adenoviral-mediated gene transfer augments antitumor immunity in patients
with metastatic melanoma. J Clin Oncol 2003;21:3343-50.
16. Luiten RM, Kueter EW, Mooi W, et al. Immunogenicity, including vitiligo, and
feasibility of vaccination with autologous GM-CSF-transduced tumor cells in metastatic
melanoma patients. J Clin Oncol 2005;23:8978-91.
15
Clavreul et al.
17. Borrello I, Sotomayor EM, Cooke S, et al. A universal granulocyte-macrophage
colony-stimulating factor-producing bystander cell line for use in the formulation of
autologous tumor cell-based vaccines. Hum Gene Ther 1999;10:1983-91.
18. Jean WC, Spellman SR, Wallenfriedman MA, et al. Effects of combined
granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2, and
interleukin-12 based immunotherapy against intracranial glioma in the rat. J Neurooncol
2004;66:39-49.
19. Clavreul A, Delhaye M, Jadaud E, et al. Effects of syngeneic cellular vaccinations
alone or in combination with GM-CSF on the weakly immunogenic F98 glioma model.
J Neurooncol 2006;79:9-17.
20. Macdonald DR, Cascino TL, Schold SC, Jr., et al. Response criteria for phase II
studies of supratentorial malignant glioma. J Clin Oncol 1990;8:1277-80.
21. Kaplan G, Walsh G, Guido LS, et al. Novel responses of human skin to intradermal
recombinant granulocyte/macrophage-colony-stimulating factor: Langerhans cell
recruitment, keratinocyte growth, and enhanced wound healing. J Exp Med
1992;175:1717-28.
22. Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and
efficacy of intraoperative controlled delivery by biodegradable polymers of
chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group.
Lancet 1995;345:1008-12.
23. De Vleeschouwer S, Fieuws S, Rutkowski S, et al. Postoperative adjuvant dendritic
cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin
Cancer Res 2008;14:3098-104.
16
Clavreul et al.
24. Kikuchi T, Akasaki Y, Irie M, et al. Results of a phase I clinical trial of vaccination
of glioma patients with fusions of dendritic and glioma cells. Cancer Immunol
Immunother 2001;50:337-44.
25. Schneider T, Gerhards R, Kirches E, et al. Preliminary results of active specific
immunization with modified tumor cell vaccine in glioblastoma multiforme. J
Neurooncol 2001;53:39-46.
26. Clavreul A, Jean I, Preisser L, et al. Human glioma cell culture: two FCS-free media
could be recommended for clinical use in immunotherapy. In Vitro Cell Dev Biol Anim
2009.
27. Wood GW, Holladay FP, Turner T, et al. A pilot study of autologous cancer cell
vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in
patients with recurrent grade III/IV astrocytoma. J Neurooncol 2000;48:113-20.
28. Ishikawa E, Tsuboi K, Yamamoto T, et al. Clinical trial of autologous formalinfixed tumor vaccine for glioblastoma multiforme patients. Cancer Sci 2007;98:1226-33.
29. Bigner DD, Pitts OM, Wikstrand CJ. Induction of lethal experimental allergic
encephalomyelitis in nonhuman primates and guinea pigs with human glioblastoma
multiforme tissue. J Neurosurg 1981;55:32-42.
30. Fazekas F, Barkhof F, Filippi M, et al. The contribution of magnetic resonance
imaging to the diagnosis of multiple sclerosis. Neurology 1999;53:448-56.
31. Yajima N, Yamanaka R, Mine T, et al. Immunologic evaluation of personalized
peptide vaccination for patients with advanced malignant glioma. Clin Cancer Res
2005;11:5900-11.
17
Clavreul et al.
32. Izumoto S, Tsuboi A, Oka Y, et al. Phase II clinical trial of Wilms tumor 1 peptide
vaccination for patients with recurrent glioblastoma multiforme. J Neurosurg
2008;108:963-71.
33. Stathopoulos A, Samuelson C, Milbouw G, et al. Therapeutic vaccination against
malignant gliomas based on allorecognition and syngeneic tumor antigens: proof of
principle in two strains of rat. Vaccine 2008;26:1764-72.
34. Mahaley MS, Jr., Bigner DD, Dudka LF, et al. Immunobiology of primary
intracranial tumors. Part 7: Active immunization of patients with anaplastic human
glioma cells: a pilot study. J Neurosurg 1983;59:201-7.
35. Liau LM, Prins RM, Kiertscher SM, et al. Dendritic cell vaccination in glioblastoma
patients induces systemic and intracranial T-cell responses modulated by the local
central nervous system tumor microenvironment. Clin Cancer Res 2005;11:5515-25.
18
Clavreul et al.
Legends of figures
Figure 1: Vaccination schedules.
Figure 2: Cutaneous biopsy before vaccination in patient n°5 showed a mild
perisvascular lymphocytic infiltration in the superficial dermis (A, original
magnification 200) and only rare CD1a positive Langerhans cells (B, original
magnification 400). After vaccination, there was an increase in perivascular T
lymphocytes (C, original magnification 200) and in CD1a positive Langerhans cells
(D, original magnification 400).
Figure 3: MRI from patients n°1, n°2, n°4, n°5 and n°8, before surgery (A, D, H, L, P),
24 h after surgery (B, E, I, M, Q), 24 h before the first vaccination (C, F, J, N, R) and 8
weeks after the final vaccination (G, K, O, S).
19