Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 61(2): 239-247, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22080408

ABSTRACT

Monitoring of immunotherapeutic clinical trials has undergone a considerable change in the last decade resulting in a general agreement that immune monitoring should guide the development of cancer vaccines. The emphasis on immune cell functions and quantitation of antigen-specific T cells have been playing a major role in the attempts to establish meaningful correlations between therapy-induced alterations in immune responses and clinical endpoints. However, one significant unresolved issue in modern immunotherapy is that when a tumor-specific cellular immune response is observed following the course of immunotherapy, it does not always lead to clinically proven cancer regression. This disappointing lack of a correlation between the tumor-specific cytotoxic immune responses and the clinical efficacy of immunotherapy may be explained, among other reasons, by the notion that the analysis of any single immunological parameter is not sufficient to provide clinically feasible information about the complex interactions between different cell subsets in the peripheral blood and immune, tumor, and stromal cells in the tumor milieu. By contrast, a systemic approach is required for improving the quality of a serial monitoring to ensure that it adequately and reliably measures potential changes induced in patients by administered vaccines or immunomodulators. Comprehensive evaluation of the balance between the immunostimulatory and immunosuppressive compartments of the immune system could be critical for a better understanding of how a given immunotherapy works or does not work in a particular clinical trial. New approaches to characterize tumor-infiltrating leukocytes, their phenotypic, biochemical, and genetic characteristics within the tumor microenvironment need to be developed and validated and should complement current monitoring techniques. These immune-monitoring assays for the local tumor immunoenvironment should be developed, validated, and standardized for reliability and consistency in order to establish the overall performance standards.


Subject(s)
Cancer Vaccines , Immunotherapy , Monitoring, Immunologic , Neoplasms/drug therapy , Neoplasms/immunology , Clinical Trials as Topic , Humans , Monitoring, Immunologic/trends , Treatment Outcome , Tumor Microenvironment
2.
Cells ; 1(2): 111-26, 2012 May 10.
Article in English | MEDLINE | ID: mdl-24710418

ABSTRACT

The profiling and monitoring of immune responses are key elements in the evaluation of the efficacy and development of new biotherapies, and a number of assays have been introduced for analyzing various immune parameters before, during, and after immunotherapy. The choice of immune assays for a given clinical trial depends on the known or suggested immunomodulating mechanisms associated with the tested therapeutic modality. Cell-mediated cytotoxicity represents a key mechanism in the immune response to various pathogens and tumors. Therefore, the selection of monitoring methods for the appropriate assessment of cell-mediated cytotoxicity is thought to be crucial. Assays that can detect both cytotoxic T lymphocytes (CTL) frequency and function, such as the IFN-γ enzyme-linked immunospot assay (ELISPOT) have gained increasing popularity for monitoring clinical trials and in basic research. Results from various clinical trials, including peptide and whole tumor cell vaccination and cytokine treatment, have shown the suitability of the IFN-γ ELISPOT assay for monitoring T cell responses. However, the Granzyme B ELISPOT assay and Perforin ELISPOT assay may represent a more direct analysis of cell-mediated cytotoxicity as compared to the IFN-γ ELISPOT, since Granzyme B and perforin are the key mediators of target cell death via the granule-mediated pathway. In this review we analyze our own data and the data reported by others with regard to the application of various modifications of ELISPOT assays for monitoring CTL activity in clinical vaccine trials.

3.
Immunotherapy ; 3(10): 1131-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21995567

ABSTRACT

Biotherapy is widely considered as the fourth treatment modality for patients with cancer, and uses the constantly increasing knowledge in molecular biology, cell biology and immunology. Biotherapy uses naturally occurring biological molecules (e.g., cytokines and antibodies) or works by the manipulation of normal biological mechanisms (controlling or inhibiting tumor growth). Important achievements in anticancer drug development are immunotherapeutic strategies recently approved by the US FDA as well as clinical data of the cancer patients treated in clinical trials. There is a need to expand these novel cancer immunotherapeutic modalities for cancer patients all over the world. To meet that goal, it is essential to spread the information, to summarize the new clinical data and to draw the conclusions from the clinical and preclinical investigations. These frontline tasks can be well advanced by organizing international conferences in this domain in less scientifically developed countries, with a significant tumor burden statistics. Therefore, special efforts were done to organize the 2nd International Cancer Immunotherapy and Immunomonitoring Conference (CITIM-2011) in Hungary.


Subject(s)
Immunotherapy , Neoplasms/immunology , Translational Research, Biomedical , Animals , Clinical Trials as Topic , Congresses as Topic , Drug Approval , Evidence-Based Medicine , Humans , Hungary , Immunotherapy/trends , Information Dissemination , Monitoring, Immunologic/trends , Neoplasms/pathology , Neoplasms/therapy
4.
Expert Opin Biol Ther ; 10(11): 1539-53, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20955111

ABSTRACT

IMPORTANCE OF THE FIELD: Dendritic cells (DC) are powerful antigen-presenting cells that induce and maintain primary cytotoxic T lymphocyte (CTL) responses directed against tumor antigens. Consequently, there has been much interest in their application as antitumor vaccines. AREAS COVERED IN THIS REVIEW: A large number of DC-based vaccine trials targeting a variety of cancers have been conducted; however, the rate of reported clinically significant responses remains low. Modification of DC to express tumor antigens or immunostimulatory molecules through the transfer of genes or mRNA transfection offers a logical alternative with potential advantages over peptide- or protein antigen-loaded DC. In this article, we review the current results and future prospects for genetically modified DC vaccines for the treatment of cancer. WHAT THE READER WILL GAIN: Genetically-modified dendritic cell-based vaccines represent a powerful tool for cancer therapy. Numerous preclinical and clinical studies have demonstrated the potential of dendritic cell vaccines alone or in combination with other therapeutic modalities. TAKE HOME MESSAGE: Genetically modified DC-based anti-cancer vaccination holds promise, perhaps being best employed in the adjuvant setting with minimal residual disease after primary therapy, or in combination with other antitumor or immune-enhancing therapies.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Gene Transfer Techniques , Immunotherapy/methods , Neoplasms/therapy , Animals , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Clinical Trials as Topic , Combined Modality Therapy , Cytokines/administration & dosage , Cytokines/genetics , DNA, Recombinant/administration & dosage , DNA, Recombinant/genetics , Dendritic Cells/immunology , Female , Humans , Killer Cells, Natural/immunology , Male , Mice , Neoplasm Proteins/administration & dosage , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/immunology , RNA Interference , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Neoplasm/administration & dosage , RNA, Neoplasm/genetics , RNA, Neoplasm/immunology , Randomized Controlled Trials as Topic , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome , Vaccination
5.
Expert Rev Vaccines ; 9(6): 601-16, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20518716

ABSTRACT

The exact immunologic responses after vaccination that result in effective antitumor immunity have not yet been fully elucidated and the data from ex vivo T-cell assays have not yet defined adequate surrogate markers for clinical efficacy. A more detailed knowledge of the specific immune responses that correlate with positive clinical outcomes should help to develop better or novel strategies to effectively activate the immune system against tumors. Furthermore, clinically relevant material is often limited and, thus, precludes the ability to perform multiple assays. The two main assays currently used to monitor lymphocyte-mediated cytoxicity in cancer patients are the (51)Cr-release assay and IFN-gamma ELISpot assay. The former has a number of disadvantages, including low sensitivity, poor labeling and high spontaneous release of isotope from some tumor target cells. Additional problems with the (51)Cr-release assay include difficulty in obtaining autologous tumor targets, and biohazard and disposal problems for the isotope. The ELISpot assays do not directly measure cytotoxic activity and are, therefore, a surrogate marker of cyotoxic capacity of effector T cells. Furthermore, they do not assess cytotoxicity mediated by the production of the TNF family of death ligands by the cytotoxic cells. Therefore, assays that allow for the simultaneous measurement of several parameters may be more advantageous for clinical monitoring. In this respect, multifactor flow cytometry-based assays are a valid addition to the currently available immunologic monitoring assays. Use of these assays will enable detection and enumeration of tumor-specific cytotoxic T lymphocytes and their specific effector functions and any correlations with clinical responses. Comprehensive, multifactor analysis of effector cell responses after vaccination may help to detect factors that determine the success or failure of a vaccine and its immunological potency.


Subject(s)
Cancer Vaccines/immunology , Cytotoxicity, Immunologic , Flow Cytometry/methods , Animals , Apoptosis , Humans , Lysosomal-Associated Membrane Protein 1/analysis , T-Lymphocytes, Cytotoxic/immunology , Vaccination
6.
J Interferon Cytokine Res ; 26(10): 706-18, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17032165

ABSTRACT

The proinflammatory cytokine, interleukin-18 (IL-18), is a natural killer (NK) cell activator that induces NK cell cytotoxicity and interferon-gamma (IFN-gamma) expression. In this report, we define a novel role for IL-18 as an NK cell protective agent. Specifically, IL-18 prevents NK cell death initiated by different and distinct stress mechanisms. IL-18 reduces NK cell self-destruction during NK-targeted cell killing, and in the presence of staurosporin, a potent apoptotic inducer, IL-18 reduces caspase-3 activity. The critical regulatory step in this process is downstream of the mitochondrion and involves reduced cleavage and activation of caspase-9 and caspase-3. The ability of IL-18 to regulate cell survival is not limited to a caspase death pathway in that IL-18 augments tumor necrosis factor (TNF) signaling, resulting in increased and prolonged mRNA expression of c-apoptosis inhibitor 2 (cIAP2), a prosurvival factor and caspase-3 inhibitor, and TNF receptor-associated factor 1 (TRAF1), a prosurvival protein. The cumulative effects of IL-18 define a novel role for this cytokine as a molecular survival switch that functions to both decrease cell death through inhibition of the mitochondrial apoptotic pathway and enhance TNF induction of prosurvival factors.


Subject(s)
Apoptosis , Inhibitor of Apoptosis Proteins/biosynthesis , Interleukin-18/pharmacology , Killer Cells, Natural/immunology , Signal Transduction , Cells, Cultured , Humans , Inflammation Mediators/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Killer Cells, Natural/drug effects , NF-kappa B/metabolism , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 1/biosynthesis , TNF Receptor-Associated Factor 1/genetics , Tumor Necrosis Factor-alpha/pharmacology
7.
J Immunol ; 170(5): 2727-33, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12594303

ABSTRACT

In cancer, the coordinate engagement of professional APC and Ag-specific cell-mediated effector cells may be vital for the induction of effective antitumor responses. We speculated that the enhanced differentiation and function of dendritic cells through CD40 engagement combined with IL-2 administration to stimulate T cell expansion would act coordinately to enhance the adaptive immune response against cancer. In mice bearing orthotopic metastatic renal cell carcinoma, only the combination of an agonist Ab to CD40 and IL-2, but neither agent administered alone, induced complete regression of metastatic tumor and specific immunity to subsequent rechallenge in the majority of treated mice. The combination of anti-CD40 and IL-2 resulted in significant increases in dendritic cell and CD8(+) T cell number in advanced tumor-bearing mice compared with either agent administered singly. The antitumor effects of anti-CD40 and IL-2 were found to be dependent on CD8(+) T cells, IFN-gamma, IL-12 p40, and Fas ligand. CD40 stimulation and IL-2 may therefore be of use to promote antitumor responses in advanced metastatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Immune Sera/administration & dosage , Interleukin-2/administration & dosage , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Adjuvants, Immunologic/administration & dosage , Animals , CD40 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Lewis Lung , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , Cell Differentiation/immunology , Dendritic Cells/pathology , Drug Synergism , Epitopes, T-Lymphocyte/immunology , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Neoplasm Metastasis/prevention & control , Neoplasm Transplantation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...