Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 62(9): 1499-509, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23817721

ABSTRACT

BACKGROUND: The growth and recurrence of several cancers appear to be driven by a population of cancer stem cells (CSCs). Glioblastoma, the most common primary brain tumor, is invariably fatal, with a median survival of approximately 1 year. Although experimental data have suggested the importance of CSCs, few data exist regarding the potential relevance and importance of these cells in a clinical setting. METHODS: We here present the first seven patients treated with a dendritic cell (DC)-based vaccine targeting CSCs in a solid tumor. Brain tumor biopsies were dissociated into single-cell suspensions, and autologous CSCs were expanded in vitro as tumorspheres. From these, CSC-mRNA was amplified and transfected into monocyte-derived autologous DCs. The DCs were aliquoted to 9-18 vaccines containing 10(7) cells each. These vaccines were injected intradermally at specified intervals after the patients had received a standard 6-week course of post-operative radio-chemotherapy. The study was registered with the ClinicalTrials.gov identifier NCT00846456. RESULTS: Autologous CSC cultures were established from ten out of eleven tumors. High-quality RNA was isolated, and mRNA was amplified in all cases. Seven patients were able to be weaned from corticosteroids to receive DC immunotherapy. An immune response induced by vaccination was identified in all seven patients. No patients developed adverse autoimmune events or other side effects. Compared to matched controls, progression-free survival was 2.9 times longer in vaccinated patients (median 694 vs. 236 days, p = 0.0018, log-rank test). CONCLUSION: These findings suggest that vaccination against glioblastoma stem cells is safe, well-tolerated, and may prolong progression-free survival.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Neoplastic Stem Cells/immunology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cancer Vaccines/immunology , Combined Modality Therapy , Dendritic Cells/pathology , Disease-Free Survival , Female , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , Telomerase/genetics , Telomerase/immunology , Transfection
2.
Oncoimmunology ; 1(5): 670-686, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22934259

ABSTRACT

Understanding the basis of a successful clinical response after treatment with therapeutic cancer vaccines is essential for the development of more efficacious therapy. After vaccination with the single telomerase (hTERT) 16-mer peptide, GV1001, some patients experienced clinical responses and long-term survival. This study reports in-depth immunological analysis of the T-cell response against telomerase (hTERT) in clinically responding patients compared with clinical non-responders following vaccination with the single hTERT 16-mer peptide, GV1001. Extensive characterization of CD4+ T-cell clones specific for GV1001 generated from a lung cancer patient in complete remission after vaccination demonstrated a very broad immune response to this single peptide vaccine with differences in fine specificity, HLA restriction, affinity and function. Some CD4+ T-cell clones were cytotoxic against peptide-loaded target cells and also recognized processed recombinant hTERT protein. Furthermore, T-cell responses against several unrelated hTERT epitopes, some of which are novel, were detected, indicating extensive epitope spreading which was confirmed in other clinical responders. In contrast, patients responding immunologically, but not clinically, after vaccination did not display this intramolecular epitope spreading. Multifunctional CD4+ T-cell clones specific for novel hTERT epitopes were generated and shown to recognize a melanoma cell line. Pentamer analysis of T cells in peripheral blood also demonstrated the presence of an important CD8+ T-cell response recognizing an HLA-B7 epitope embedded in GV1001 not previously described. These results indicate that the highly diverse hTERT-specific T-cell response, integrating both T helper and CTL responses, is essential for tumor regression and the generation of long-term T-cell memory.

3.
Cancer Immunol Immunother ; 60(11): 1553-64, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21681371

ABSTRACT

PURPOSE: A phase I study was conducted to investigate the safety, tolerability, and immunological responses to vaccination with a combination of telomerase-derived peptides GV1001 (hTERT: 611-626) and p540 (hTERT: 540-548) using granulocyte-macrophage colony-stimulating factor (GM-CSF) or tuberculin as adjuvant in patients with cutaneous melanoma. EXPERIMENTAL DESIGN: Ten patients with melanoma stages UICC IIb-IV were vaccinated 8 times intradermally with either 60 or 300 nmole of GV1001 and p540 peptide using GM-CSF as adjuvant. A second group of patients received only 300 nmole GV1001 in combination with tuberculin PPD23 injections. HLA typing was not used as an inclusion criterion. Peptide-specific immune responses were measured by delayed-type hypersensitivity (DTH) reactions, in vitro T cell proliferation assays, and cytotoxicity (51-Chromium release) assays for a selected number of clones subsequently generated. RESULTS: Vaccination was well tolerated in all patients. Peptide-specific immune response measured by DTH reactions and in vitro response could be induced in a dose-dependent fashion in 7 of 10 patients. Cloned T cells from the vaccinated patients showed proliferative responses against both vaccine peptides GV1001 and p540. Furthermore, T cell clones were able to specifically lyse p540-pulsed T2 target cells and various pulsed and unpulsed tumor cell lines. CONCLUSION: These results demonstrate that immunity to hTERT can be generated safely and effectively in patients with advanced melanoma and therefore encourage further trials.


Subject(s)
Cancer Vaccines/therapeutic use , Melanoma/therapy , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , Skin Neoplasms/drug therapy , Telomerase/immunology , Telomerase/therapeutic use , Adult , Aged , Amino Acid Sequence , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Female , Humans , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Molecular Sequence Data , Peptide Fragments/adverse effects , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Telomerase/adverse effects
4.
Cancer Immunol Immunother ; 60(6): 809-18, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21365467

ABSTRACT

Immunotherapy targeting the hTERT subunit of telomerase has been shown to induce robust immune responses in cancer patients after vaccination with single hTERT peptides. Vaccination with dendritic cells (DCs) transfected with hTERT mRNA has the potential to induce strong immune responses to multiple hTERT epitopes and is therefore an attractive approach to more potent immunotherapy. Blood samples from such patients provide an opportunity for identification of new, in vivo processed T-cell epitopes that may be clinically relevant. A 62-year-old female patient underwent radical surgery for a pancreatic adenocarcinoma. After relapse, she obtained stable disease on gemcitabine treatment. Due to severe neutropenia, the chemotherapy was terminated. The patient has subsequently been treated with autologous DCs loaded with hTERT mRNA for 3 years. Immunomonitoring was performed at regular intervals following start of vaccination and clinical outcome measured by CT and PET/CT evaluation. The patient developed an immune response against several hTERT-derived Th and CTL epitopes. She presently shows no evidence of active disease based on PET/CT scans. No serious adverse events were experienced and the patient continues to receive regular booster injections. We here provide evidence for the induction of hTERT-specific immune responses following vaccination of a pancreas cancer patient with DCs loaded with hTERT mRNA. These responses are associated with complete remission. A thorough analysis of this patient immune response has provided a unique opportunity to identify novel epitopes, associated with clinical effects. These will be included in future hTERT vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Dendritic Cells/immunology , Pancreatic Neoplasms/therapy , RNA, Messenger/immunology , Telomerase/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Growth Processes/immunology , Cell Line, Tumor , Epitopes/genetics , Epitopes/immunology , Female , Humans , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Telomerase/genetics , Transfection
5.
Cytotherapy ; 13(5): 629-40, 2011 May.
Article in English | MEDLINE | ID: mdl-21174490

ABSTRACT

BACKGROUND AIMS: T cells can be redirected to reject cancer by retroviral transduction with a chimeric antigen receptor (CAR) or by administration of a bispecific T cell engager (BiTE). We demonstrate that transfection of T cells with messenger (m) RNA coding for CAR is an alternative strategy. METHODS: We describe the pre-clinical evaluation of a method based on transient modification of expanded T cells with a CD19 CAR directed against B-cell malignancies. CAR mRNA was generated under cell-free conditions in a scalable process using recombinant RNA polymerase. Efficient and non-toxic square-wave electroporation was used to load the mRNA into the cytoplasm of T cells with no risk of insertional mutagenesis. RESULTS: After transfection >80% of T cells were viable, with 94% CAR expression. Transfected T cells were cytolytic to CD19(+) targets and produced interferon (IFN)-γ in response. Killing of CD19(+) target cells was demonstrated even at day 8 with undetectable CAR expression. Increasing the concentration of mRNA resulted in higher surface CAR expression, better killing and more IFN-γ release but at the expense of increased activation-induced cell death. Finally, we demonstrated that a second transgene could be introduced by co-electroporation of CXCR4 or CCR7 with CAR to also modify chemotactic responses. CONCLUSIONS: We advocate the transient redirection approach as well suited to meet safety aspects for early phase studies, prior to trials using stably transduced cells once CAR has been proven safe. The simplicity of this methodology also facilitates rapid screening of candidate targets and novel receptors in pre-clinical studies.


Subject(s)
Adoptive Transfer/methods , Leukemia/therapy , Lymphoma/therapy , Receptors, Antigen/genetics , Receptors, CCR7/genetics , Receptors, CXCR4/genetics , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Antigens, CD19/analysis , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Retroviridae , Transduction, Genetic
6.
Methods Mol Biol ; 629: 453-93, 2010.
Article in English | MEDLINE | ID: mdl-20387166

ABSTRACT

Adoptive transfer of T cells with restricted tumor specificity provides a promising approach to immunotherapy of cancers. However, the isolation of autologous cytotoxic T cells that recognize tumor-associated antigens is time consuming and fails in many instances. Alternatively, gene modification with tumor antigen-specific T-cell receptors (TCR) or chimeric antigen receptors (CARs) can be used to redirect the specificity of large numbers of immune cells toward the malignant cells. Chimeric antigen receptors are composed of the single-chain variable fragment (scFv) of a tumor-recognizing antibody cloned in frame with human T-cell signaling domains (e.g., CD3zeta, CD28, OX40, 4-1BB), thus combining the specificity of antibodies with the effector functions of cytotoxic T cells. Upon antigen binding, the intracellular signaling domains of the CAR initiate cellular activation mechanisms including cytokine secretion and cytolysis of the antigen-positive target cell.In this chapter, we provide detailed protocols for large-scale ex vivo expansion of T cells and manufacturing of medium-scale batches of CAR-expressing T cells for translational research by mRNA electroporation. An anti-CD19 chimeric receptor for the targeting of leukemias and lymphomas was used as a model system. We are currently scaling up the protocols to adapt them to cGMP production of a large number of redirected T cells for clinical applications.


Subject(s)
Cell Culture Techniques/methods , HLA Antigens/immunology , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Cell Proliferation , Cell Separation , Cell Survival , Cryopreservation , Cytotoxicity, Immunologic , Humans , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Lymphocyte Depletion , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Immunologic , Reproducibility of Results , Transfection
7.
J Immunol Methods ; 355(1-2): 52-60, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20171968

ABSTRACT

Adoptive T cell therapy is a promising treatment strategy for patients with different types of cancer. The methods used for generation of high numbers of tumor specific T cells usually require long-term ex vivo culture, which frequently lead to generation of terminally differentiated effector cells, demonstrating low persistence in vivo. Therefore, optimization of protocols for generation of T cells for adoptive cell therapy is warranted. The aim of this work was to develop a protocol for expansion of antigen-specific T cells using Dynabeads CD3/CD28 to obtain T cells expressing markers important for in vivo persistence and survival. To achieve high numbers of antigen-specific T cells following expansion, we have tested the effect of depleting regulatory T cells using Dynabeads CD25 and including a pre-stimulation step with peptide prior to the non-specific expansion with Dynabeads. Our data demonstrate that virus- and tumor specific T cells can be expanded to high numbers using Dynabeads CD3/CD28 following optimization of the culture conditions. The expansion protocol presented here results in enrichment of antigen-specific CD8(+) T cells with an early/intermediate memory phenotype. This is observed even when the antigen-specific CD8(+) T cells demonstrated a terminal effector phenotype prior to expansion. This protocol thus results in expanded T cells with a phenotypic profile which may increase the chance of retaining long-term persistence following adoptive transfer. Based on these data we have developed a cGMP protocol for expansion of tumor specific T cells for adoptive T cell therapy.


Subject(s)
Adoptive Transfer , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunomagnetic Separation , Cell Culture Techniques/methods , Cell Survival , Female , Humans , Immunologic Memory , Male , Neoplasms/immunology , Neoplasms/therapy
8.
J Med Entomol ; 43(3): 533-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16739412

ABSTRACT

Head lice, Pediculis capitis De Geer, populations were investigated for permethrin and malathion resistance after initial establishment of a discriminating dose of topical application bioassay with body lice, Pediculus humanus L. For both insecticides, approximately 2 times the lethal dose (LD)95 at 4 h was selected, 2 ng of permethrin and 100 ng of malathion per head louse, respectively. Head lice were collected from heads of infested children in Denmark at 33 primary schools, one kindergarten, and seven boarding schools. The lice were collected by combing of dry hair, with a fine-toothed antilouse comb attached to a vacuum cleaner. A resistance survey covers head lice collected from 208 of 1,441 persons combed. The frequency of permethrin- and malathion-resistant head lice is high in Danish head lice populations. In 17 of 24 samples tested for permethrin resistance, all head lice survived the discriminating dose. Six samples had between 3 and 25% dead head lice, whereas one sample had 60% mortality. In nine of 25 samples tested for malathion resistance, all head lice survived the discriminating dose. Seven samples had <25% dead head lice, and four samples had a mortality of 50% or more at the discriminating dose. The connection between permethrin resistance and kdr-like mutations is confirmed by our findings. The frequency of the double mutation T929I-L932 F in the voltage-sensitive sodium channel gene associated with permethrin resistance was 0.95 in Danish head lice populations.


Subject(s)
Insecticide Resistance , Malathion , Pediculus , Permethrin , Adolescent , Adult , Animals , Child , Child, Preschool , Denmark , Humans , Insecticide Resistance/genetics , Lice Infestations/drug therapy , Malathion/administration & dosage , Mutation , Pediculus/genetics , Permethrin/administration & dosage , Sodium Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...