Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 624685, 2021.
Article in English | MEDLINE | ID: mdl-33679769

ABSTRACT

Antigen-specific therapy for multiple sclerosis may lead to a more effective therapy by induction of tolerance to a wide range of myelin-derived antigens without hampering the normal surveillance and effector function of the immune system. Numerous attempts to restore tolerance toward myelin-derived antigens have been made over the past decades, both in animal models of multiple sclerosis and in clinical trials for multiple sclerosis patients. In this review, we will give an overview of the current approaches for antigen-specific therapy that are in clinical development for multiple sclerosis as well provide an insight into the challenges for future antigen-specific treatment strategies for multiple sclerosis.


Subject(s)
Adoptive Transfer , Desensitization, Immunologic , Multiple Sclerosis/therapy , Myelin Proteins/administration & dosage , Peptide Fragments/administration & dosage , Vaccination , Vaccines/therapeutic use , Adoptive Transfer/adverse effects , Adoptive Transfer/history , Adoptive Transfer/trends , Animals , Autoimmunity , Desensitization, Immunologic/adverse effects , Desensitization, Immunologic/history , Desensitization, Immunologic/trends , Diffusion of Innovation , Forecasting , History, 20th Century , History, 21st Century , Humans , Immune Tolerance , Multiple Sclerosis/history , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Myelin Proteins/adverse effects , Myelin Proteins/immunology , Myelin Proteins/metabolism , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Peptide Fragments/metabolism , Vaccination/adverse effects , Vaccination/history , Vaccination/trends , Vaccines/adverse effects
2.
Trends Mol Med ; 25(11): 980-992, 2019 11.
Article in English | MEDLINE | ID: mdl-31494024

ABSTRACT

Cancer immunotherapy includes cancer vaccination, adoptive T cell transfer (ACT) with chimeric antigen receptor (CAR) T cells, and administration of tumor-infiltrating lymphocytes and immune-checkpoint blockade such as anti-CTLA4/anti-PD1 inhibitors that can directly or indirectly target tumor neoantigens and elicit a T cell response. Accurate, rapid, and cost-effective identification of neoantigens, however, is critical for successful immunotherapy. Here, we review computational issues for neoantigen identification by summarizing the various sources of neoantigens and their identification from high-throughput sequencing data. Several opinions are presented to inspire further discussions toward improving neoantigen identification. Continuing efforts are required to improve the sensitivity and specificity of bona fide neoantigens, taking advantage of the development of high-throughput sequencing techniques for effective and personalized cancer immunotherapy.


Subject(s)
Antigens, Neoplasm , Immunotherapy/trends , Neoplasms/immunology , Adoptive Transfer/trends , Alternative Splicing , Antigens, Neoplasm/genetics , Antigens, Neoplasm/isolation & purification , Biomarkers, Tumor , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Computer Simulation/trends , Gene Expression Profiling , Genomics , High-Throughput Nucleotide Sequencing , Humans , Immunotherapy, Adoptive/trends , Loss of Heterozygosity , Major Histocompatibility Complex/genetics , Neoplasms/therapy , Prognosis , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Proteomics , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
3.
Transplantation ; 101(10): 2277-2287, 2017 10.
Article in English | MEDLINE | ID: mdl-28376037

ABSTRACT

Cellular therapy with CD4FOXP3 T regulatory (Treg) cells is a promising strategy to induce tolerance after solid-organ transplantation or prevent graft-versus-host disease after transfer of hematopoietic stem cells. Treg cells currently used in clinical trials are either polyclonal, donor- or antigen-specific. Aside from variations in isolation and expansion protocols, however, most therapeutic Treg cell-based products are much alike. Ongoing basic science work has provided considerable new insight into multiple facets of Treg cell biology, including their stability, homing, and functional specialization; integrating these basic science discoveries with clinical efforts will support the development of next-generation therapeutic Treg cells with enhanced efficacy. In this review, we summarize recent advances in knowledge of how Treg cells home to lymphoid and peripheral tissues, and control antibody production and tissue repair. We also discuss newly appreciated pathways that modulate context-specific Treg cell function and stability. Strategies to improve and tailor Treg cells for cell therapy to induce transplantation tolerance are highlighted.


Subject(s)
Adoptive Transfer/trends , Allergy and Immunology/trends , Graft Rejection/prevention & control , Organ Transplantation/trends , T-Lymphocytes, Regulatory/transplantation , Animals , B-Lymphocytes/immunology , Chemotaxis, Leukocyte , Diffusion of Innovation , Forecasting , Genetic Therapy/methods , Graft Rejection/immunology , Graft Survival , Humans , Organ Transplantation/adverse effects , Phenotype , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transplantation Tolerance , Treatment Outcome
4.
Discov Med ; 22(121): 215-220, 2016 10.
Article in English | MEDLINE | ID: mdl-27875673

ABSTRACT

Immunotherapy using engineered autologous T cells has been attempted for decades, but clinical trials have only recently demonstrated efficacy. The combination of enhanced manufacturing techniques, highly efficient engineering, appropriate target selection and synthetic receptors with potent T cell activating domains has led to the development of highly-active cellular therapy products. B-cell malignancies have served as the paradigmatic diseases to initially evaluate and subsequently hone engineered T cells targeting cancer. Two engineered receptors, transgenic T cell receptors (tTCRs) and chimeric antigen receptors (CARs), have been explored clinically at several different institutions. The most profound success has been in pediatric and adult acute lymphoblastic leukemia, in which complete response rates after treatment with CD19-directed CAR T cells approach 90%. Success has been slightly less impressive in slower-growing diseases such as chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL), and experience is much more limited in the plasma cell disease multiple myeloma. A great deal of investigation is underway to understand the differences in response rates observed, and enhance the efficacy of these therapies in B cell cancers. Here, we review landmark and recent clinical trials, as well as pre-clinical work that demonstrates significant promise in propelling this field further in the coming years.


Subject(s)
Adoptive Transfer/methods , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen , Recombinant Fusion Proteins , T-Lymphocytes/transplantation , Adoptive Transfer/trends , Female , Humans , Male
5.
Crit Rev Oncog ; 20(3-4): 315-27, 2015.
Article in English | MEDLINE | ID: mdl-26349422

ABSTRACT

After decades of basic research, immune-based therapeutics for the treatment of cancer are showing evidence of efficacy in clinical trials; several immunotherapeutics already incorporated into standard treatment regimens. Intensive research is underway to improve the efficacy of immunotherapeutics and to expand the application of immunotherapy to a wider array of cancers. The therapeutic options that comprise immunotherapy for cancer are vast and span monoclonal antibodies, tumor vaccines, adoptive cellular therapies, as well as therapies aimed at reversing immunosuppression and enhancing immune reactivity globally and/or locally within the tumor microenvironment. In pediatric cancer, monoclonal antibodies have demonstrated efficacy in hematologic malignancies, and neuroblastoma and bispecific antibodies that activate resident T cells, as well as adoptive cell therapy, have shown recent exciting results for the treatment of acute lymphoblastic leukemia in childhood. This review discusses the basic principles of tumor immunology driving clinical development of new immunotherapies, describes immunotherapeutics with demonstrated efficacy and several currently in clinical trials, and highlight agents that seem to be most promising for the treatment of pediatric cancer.


Subject(s)
Adoptive Transfer/trends , Genetic Therapy/trends , Medical Oncology/trends , Neoplasms/immunology , Neoplasms/therapy , Adoptive Transfer/methods , Animals , Cancer Vaccines/administration & dosage , Child , Genetic Therapy/methods , Humans , Immunotherapy/methods , Immunotherapy/trends , Medical Oncology/methods , Neoplasms/genetics
8.
Expert Opin Biol Ther ; 9(4): 451-64, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19344282

ABSTRACT

BACKGROUND: Recent findings on T cells and dendritic cells have elucidated principles that can be used for a bottom-up approach to engineering artificial antigen presentation on synthetic substrates. OBJECTIVE/METHODS: To compare the latest artificial antigen-presenting cell (aAPC) technology, focussing on acellular systems because they offer advantages such as easy tunability and rapid point-of-care application compared with cellular systems. We review acellular aAPC performance and discuss their promise for clinical applications. RESULTS/CONCLUSION: Acellular aAPCs are a powerful alternative to natural-cell-based therapies, offering flexibility and modularity for incorporation oSf a variety of stimuli, hence increasing precision. Current technologies should adapt physiologically important signals within safe materials to more closely approximate their cellular counterparts. These constructs could be administered parenterally as APC replacements for active vaccines or used ex vivo for adoptive immunotherapy.


Subject(s)
Adoptive Transfer/methods , Antigen Presentation/immunology , Vaccines, Acellular/immunology , Adoptive Transfer/trends , Animals , Humans , Immunotherapy, Active/methods , Immunotherapy, Active/trends , Vaccines, Acellular/administration & dosage
9.
Expert Opin Biol Ther ; 9(5): 579-91, 2009 May.
Article in English | MEDLINE | ID: mdl-19368527

ABSTRACT

BACKGROUND: Adoptive transfer of T-lymphocytes is a promising treatment for a variety of malignancies, but is often not feasible due to difficulties in generating T-cells reactive with the targeted antigen from patients. To facilitate rapid generation of cells for therapy, T-cells can be programmed with genes encoding for an antigen-specific T-cell receptor (TCR) or chimeric receptors. OBJECTIVE: To discuss the molecular design and selected pitfalls of TCR gene modified T-cells and T-cells expressing chimeric receptors, so called T-bodies. METHODS: A selected review of the recent literature. CONCLUSION: Clinical trials report so far only limited efficacy of adoptively transferred genetically modified T-cells. However, the recent progress in engineering tumor-reactive T cells is providing a promising basis to further explore this treatment modality.


Subject(s)
Gene Targeting/methods , Gene Transfer Techniques , Neoplasms/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adoptive Transfer/methods , Adoptive Transfer/trends , Animals , Clinical Trials as Topic/methods , Clinical Trials as Topic/trends , Gene Targeting/trends , Gene Transfer Techniques/trends , Humans , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Receptors, Antigen, T-Cell/therapeutic use
12.
Expert Rev Vaccines ; 5(1): 55-65, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16451108

ABSTRACT

Recent studies have elucidated the functional links between natural killer (NK) cells and, demonstrating the reciprocal activation of these cell types through NK-DC interactions. The subsets of cells and molecular pathways involved in such interactions have been defined, and the possible anatomical sites of these interactions have also been reported. Murine experiments have demonstrated that injection of mature DCs induces rapid recruitment of NK cells to lymph nodes and that these NK cells provide interferon-gamma for Type 1 priming. Thus, there is an increasing body of in vivo evidence indicating that NK-DC interactions during the early phase of innate immunity can impact the quality and magnitude of the subsequent adaptive immune response. Importantly, these studies imply that NK cells might not serve merely as cytotoxic lymphocytes combating viral pathogens and malignant tumors, but must also be considered as important immunoregulatory cells with a significant influence on adaptive immunity. In contrast to the large volume of knowledge obtained through basic research, there is a relative paucity of information regarding NK cell function in adaptive immunity from clinical trials, as few DC vaccine studies have attempted to evaluate the nonspecific, yet potentially clinically relevant, NK response to immunization. In this article, the authors will review studies focusing on NK-DC interactions and highlight the most recent clinical findings relating to the potential role of NK cells in DC-based vaccine therapy.


Subject(s)
Adoptive Transfer/methods , Cell Communication/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Killer Cells, Natural/immunology , Adoptive Transfer/trends , Animals , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism
14.
Blood Cells Mol Dis ; 33(3): 248-55, 2004.
Article in English | MEDLINE | ID: mdl-15528139

ABSTRACT

Dendritic cells (DCs) are uniquely able to initiate and control the immune response to fungi. DCs function at three levels in the manipulation of the immune response to these pathogens. First, they mount an immediate or innate response to them, for example, by producing inflammatory mediators upon capture and phagocytosis; second, through these preceding innate functions, they decode the fungus-associated information and translate it in qualitatively different Th responses, and third, they are key in containing and dampening inflammatory responses by tolerization through the induction of regulatory T cells (Treg). DCs sense fungi in a morphotype-specific manner, through the engagement of distinct recognition receptors ultimately affecting cytokine production and costimulation. Both myeloid and plasmacytoid murine and human DCs phagocytose fungi and undergo functional maturation in response to them. However, their activation program for cytokine production was different, being IL-12 mainly produced by myeloid DCs and IL-12, IL-10 and IFN-alpha mainly produced by plasmacytoid DCs. This resulted in a distinct ability for T cell priming, being Th1, Th2, and Treg differently activated by the different DC subsets. The ability of fungus-pulsed DCs to prime for Th1 and Th2 cell activation upon adoptive transfer in vivo correlated with the occurrence of resistance and susceptibility to the infections, respectively. Antifungal protective immunity was also induced upon adoptive transfer of DCs transfected with fungal RNA. The efficacy was restricted to DCs transfected with RNA from yeasts or conidia but not with RNA from fungal hyphae. The effect was fungus-specific, as no cross-protection was observed upon adoptive transfer of DCs pulsed with either fungal species. The infusion of fungus-pulsed or RNA-transfected DCs accelerated the recovery of functional antifungal Th1 responses in mice with allogeneic hematopoietic stem cell transplantation (HSCT) and affected the outcome of the infections. As the ability of phagocytose fungi was defective in peripheral DCs from patients with HSCT, soon after the transplant, our findings suggest that the adoptive transfer of DCs may restore immunocompetence in HSCT by contributing to the educational program of T cells. Thus, the remarkable functional plasticity of DCs in response to fungi can be exploited for the deliberate targeting of cells and pathways of cell-mediated immunity in response to fungal vaccines.


Subject(s)
Adoptive Transfer , Aspergillosis/therapy , Candidiasis/therapy , Dendritic Cells/transplantation , Hematopoietic Stem Cell Transplantation , Vaccination , Adoptive Transfer/trends , Animals , Aspergillosis/immunology , Candidiasis/immunology , Dendritic Cells/immunology , Humans , Mice , Vaccination/trends
SELECTION OF CITATIONS
SEARCH DETAIL
...