Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cytometry B Clin Cytom ; 90(1): 73-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26108351

ABSTRACT

The FDA has co-sponsored three workshops to address minimal residual disease (MRD) detection in acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL), and acute myeloid leukemia (AML) as well as an FDA-NCI roundtable symposium on MRD detection and its use as a response biomarker in Multiple Myeloma (MM). As clinical outcomes in MM continue to improve with the introduction of new therapeutics, consideration of biomarkers and their development as validated surrogate endpoints that can be used in the place of traditional clinical trial endpoints of progression-free survival (PFS) will be fundamental to expeditious drug development. This article will describe the FDA drug approval process, the regulatory framework through which a biomarker can be used as a surrogate endpoint for drug approval, and how MRD detection in MM fits within this context. In parallel, this article will also describe the FDA current device clearance process with emphasis on the analytical development as it might apply to an in vitro diagnostic assay for the detection of MRD in MM. It is anticipated that this Special Issue may possibly represent how MRD might serve as a drug development tool in hematological malignancies.


Subject(s)
Antigens, CD/analysis , Antineoplastic Agents/therapeutic use , Drug Approval/legislation & jurisprudence , Flow Cytometry/standards , Multiple Myeloma/diagnosis , Neoplasm, Residual/diagnosis , Antigens, CD/genetics , Antigens, CD/immunology , Biomarkers, Pharmacological/analysis , Device Approval/legislation & jurisprudence , Gene Expression , Humans , Immunophenotyping , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Multiple Myeloma/mortality , Neoplasm, Residual/drug therapy , Neoplasm, Residual/immunology , Neoplasm, Residual/mortality , Plasma Cells/drug effects , Plasma Cells/immunology , Plasma Cells/pathology , Prognosis , Remission Induction , Survival Analysis , United States , United States Food and Drug Administration
3.
J Immunol ; 187(5): 2405-17, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21821796

ABSTRACT

Ag-specific tolerance is a highly desired therapy for immune-mediated diseases. Intravenous infusion of protein/peptide Ags linked to syngeneic splenic leukocytes with ethylene carbodiimide (Ag-coupled splenocytes [Ag-SP]) has been demonstrated to be a highly efficient method for inducing peripheral, Ag-specific T cell tolerance for treatment of autoimmune disease. However, little is understood about the mechanisms underlying this therapy. In this study, we show that apoptotic Ag-SP accumulate in the splenic marginal zone, where their uptake by F4/80(+) macrophages induces production of IL-10, which upregulates the expression of the immunomodulatory costimulatory molecule PD-L1 that is essential for Ag-SP tolerance induction. Ag-SP infusion also induces T regulatory cells that are dispensable for tolerance induction but required for long-term tolerance maintenance. Collectively, these results indicate that Ag-SP tolerance recapitulates how tolerance is normally maintained in the hematopoietic compartment and highlight the interplay between the innate and adaptive immune systems in the induction of Ag-SP tolerance. To our knowledge, we show for the first time that tolerance results from the synergistic effects of two distinct mechanisms, PD-L1-dependent T cell-intrinsic unresponsiveness and the activation of T regulatory cells. These findings are particularly relevant as this tolerance protocol is currently being tested in a Phase I/IIa clinical trial in new-onset relapsing-remitting multiple sclerosis.


Subject(s)
Immune Tolerance/immunology , Macrophages/immunology , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Apoptosis/immunology , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , B7-H1 Antigen , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunohistochemistry , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Macrophage Activation/immunology , Macrophages/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Peptides/immunology , Spleen/cytology
4.
Results Probl Cell Differ ; 51: 217-35, 2010.
Article in English | MEDLINE | ID: mdl-19130025

ABSTRACT

A primary focus in autoimmunity is the breakdown of central and peripheral tolerance resulting in the survival and eventual activation of autoreactive T cells. As CD4(+) T cells are key contributors to the underlying pathogenic mechanisms responsible for onset and progression of most autoimmune diseases, they are a logical target for therapeutic strategies. One method for restoring self-tolerance is to exploit the endogenous regulatory mechanisms that govern CD4(+) T cell activation. In this review, we discuss tolerance strategies with the common goal of inducing antigen (Ag)-specific tolerance. Emphasis is given to the use of peptide-specific tolerance strategies, focusing on ethylene carbodiimide (ECDI)-peptide-coupled cells (Ag-SP) and nonmitogenic anti-CD3, which specifically target the T cell receptor (TCR) in the absence of costimulatory signals. These approaches induce a TCR signal of insufficient strength to cause CD4(+) T cell activation and instead lead to functional T cell anergy/deletion and activation of Ag-specific induced regulatory T cells (iTregs) while avoiding generalized long-term immunosuppression.


Subject(s)
Immunosuppression Therapy/methods , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Autoantigens , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Carbodiimides , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Immunity, Mucosal , Immunosuppression Therapy/trends , Mice , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Self Tolerance , T-Lymphocytes, Regulatory/immunology
5.
Adv Exp Med Biol ; 640: 234-51, 2008.
Article in English | MEDLINE | ID: mdl-19065796

ABSTRACT

CD4+ T-cell-mediated autoimmune diseases are initiated and maintained by the presentation of self-antigen by antigen-presenting cells (APCs) to self-reactive CD4+ T-cells. According to the two-signal hypothesis, activation of a naive antigen-specific CD4+ T-cell requires stimulation of both the T-cell antigen receptor (signal 1) and costimulatory molecules such as CD28 (signal 2). To date, the majority of therapies for autoimmune diseases approved by the Food and Drug Administration primarily focus on the global inhibition of immune inflammatory activity. The goal of ongoing research in this field is to develop antigen-specific treatments which block the deleterious effects of self-reactive immune cell function while maintaining the ability of the immune system to clear nonself antigens. To this end, the signaling pathways involved in the induction of CD4+ T-cell anergy, as apposed to activation, are a topic of intense interest. This chapter discusses components of the CD4+ T-cell activation pathway that may serve as therapeutic targets for the treatment of autoimmune disease.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Animals , Humans , Immune Tolerance/immunology , Immunological Synapses/immunology
6.
Nat Rev Immunol ; 7(9): 665-77, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17690713

ABSTRACT

The development of safe and effective antigen-specific therapies is needed to treat patients with autoimmune diseases. These therapies must allow for the specific tolerization of self-reactive immune cells without altering host immunity to infectious insults. Experimental models and clinical trials for the treatment of autoimmune disease have identified putative mechanisms by which antigen-specific therapies induce tolerance. Although advances have been made in the development of efficient antigen-specific therapies, translating these therapies from bench to bedside has remained difficult. Here, we discuss the recent advances in our understanding of antigen-specific therapies for the treatment of autoimmune diseases.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/therapy , Immune Tolerance , Immunotherapy/methods , Peptides/therapeutic use , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/prevention & control , Clinical Trials as Topic , Disease Models, Animal , Humans , Mice , Peptides/immunology
7.
J Immunol ; 178(4): 2212-20, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277126

ABSTRACT

MHC class II (MHC II)-restricted T cell responses are a common driving force of autoimmune disease. Accordingly, numerous therapeutic strategies target CD4(+) T cells with the hope of attenuating autoimmune responses and restoring self-tolerance. We have previously reported that i.v. treatment with Ag-pulsed, ethylenecarbodiimide (ECDI)-fixed splenocytes (Ag-SPs) is an efficient protocol to induce Ag-specific tolerance for prevention and treatment of experimental autoimmune encephalomyelitis (EAE). Ag-SPs coupled with peptide can directly present peptide:MHC II complexes to target CD4(+) T cells in the absence of costimulation to induce anergy. However, Ag-SPs coupled with whole protein also efficiently attenuates Ag-specific T cell responses suggesting the potential contribution of alternative indirect mechanisms/interactions between the Ag-SPs and target CD4(+) T cells. Thus, we investigated whether MHC II compatibility was essential to the underlying mechanisms by which Ag-SP induces tolerance during autoimmune disease. Using MHC-deficient, allogeneic, and/or syngeneic donor Ag-SPs, we show that MHC compatibility between the Ag-SP donor and the host is not required for tolerance induction. Interestingly, we found that ECDI treatment induces apoptosis of the donor cell population which promotes uptake and reprocessing of donor cell peptides by host APCs resulting in the apparent MHC II-independent induction of tolerance. However, syngeneic donor cells are more efficient at inducing tolerance, suggesting that Ag-SPs induce functional Ag-SP tolerance via both direct and indirect (cross-tolerance) mechanisms leading to prevention and effective treatment of autoimmune disease.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance , Animals , Antigen Presentation/drug effects , Antigen-Presenting Cells/transplantation , Apoptosis/immunology , Autoantigens/immunology , Autoantigens/pharmacology , Carbodiimides/chemistry , Cell Communication/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Fixatives/chemistry , Histocompatibility Antigens Class II/immunology , Immune Tolerance/drug effects , Mice , Mice, Inbred BALB C , Transplantation, Homologous , Transplantation, Isogeneic
8.
Int Rev Immunol ; 24(5-6): 361-92, 2005.
Article in English | MEDLINE | ID: mdl-16318987

ABSTRACT

A principal theme in autoimmunity is the breakdown of central tolerance resulting in the persistence and eventual activation of autoreactive T cells. Because CD4(+) T cells are key contributors to the underlying pathogenic mechanisms responsible for the onset and progression of most autoimmune diseases, they are a logical target for therapeutic interventions. One technique for restoring self-tolerance is to exploit the endogenous regulatory mechanisms that govern CD4(+) T-cell activation. In this review, we discuss promising techniques with the common goal of inducing antigen (Ag)-specific tolerance. Emphasis is given to the use of non-mitogenic anti-CD3 and peptide-specific tolerance strategies that specifically target the T-cell receptor (TCR) in the absence of costimulatory signals. These approaches produce a TCR signal of insufficient strength to cause CD4(+) T-cell activation and instead induce functional T-cell anergy or deletion while avoiding generalized long-term immunosuppression.


Subject(s)
Autoimmune Diseases of the Nervous System/therapy , Immunotherapy/methods , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Self Tolerance/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases of the Nervous System/immunology , CD3 Complex/immunology , Clinical Trials as Topic , Cytokines/immunology , Humans , Immunosuppression Therapy/methods , Models, Immunological , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Peptides/therapeutic use , Receptors, Interleukin-2/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology
9.
Nat Immunol ; 6(7): 680-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15991363

ABSTRACT

Notch receptors are processed by gamma-secretase acting in synergy with T cell receptor signaling to sustain peripheral T cell activation. Activated CD4+ T cells differentiate into T helper type 1 (TH1) or TH2 subsets. Molecular cues directing TH1 differentiation include expression of the TH1-specific transcription factor T-bet, encoded by Tbx21. However, the regulation of Tbx21 remains incompletely defined. Here we report that Notch1 can directly regulate Tbx21 through complexes formed on the Tbx21 promoter. In vitro, gamma-secretase inhibitors extinguished expression of Notch, interferon-gamma and Tbx21 in TH1-polarized CD4+ cells, whereas ectopic expression of activated Notch1 restored Tbx21 transcription. In vivo, administration of gamma-secretase inhibitors substantially impeded TH1-mediated disease progression in the mouse experimental autoimmune encephalomyelitis model of multiple sclerosis. Thus, using gamma-secretase inhibitors to modulate Notch signaling may prove beneficial in treating TH1-mediated autoimmunity.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Endopeptidases/immunology , Protease Inhibitors/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Th1 Cells/immunology , Transcription Factors/antagonists & inhibitors , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Cytokines/immunology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/immunology , Immunoblotting , Mice , Mice, Inbred C57BL , Receptor, Notch1 , Receptors, Cell Surface/immunology , T-Box Domain Proteins , Th1 Cells/drug effects , Th1 Cells/enzymology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
10.
J Immunol ; 172(12): 7442-50, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15187122

ABSTRACT

Intravenous treatment with Ag (peptide)-coupled, ethylene carbodiimide-fixed syngeneic splenocytes (Ag-SP) is a powerful method to induce anergy in vitro and peripheral T cell tolerance in vivo. In this study, we examined the effects of Ag-SP administration on T cell activity ex vivo and in vivo using OVA-specific DO11.10 TCR transgenic T cells. Although treatment with OVA323-339-SP resulted in a strong inhibition of peptide-specific T cell recall responses in vitro, examination of the immediate effects of Ag-SP treatment on T cells in vivo demonstrated that tolerogen injection resulted in rapid T cell activation and proliferation. Although there was an increase in the number of OVA-specific DO11.10 T cells detected in the lymphoid organs, these previously tolerized T cells were strongly inhibited in mounting proliferative or inflammatory responses upon rechallenge in vivo with peptide in CFA. This unresponsiveness was reversible by treatment with anti-CTLA-4 mAb. These results are consistent with the hypothesis that Ag-SP injection induces a state of T cell anergy that is maintained by CTLA-4 engagement.


Subject(s)
Antigens, Differentiation/physiology , Immune Tolerance , Lymphocyte Activation , Th1 Cells/cytology , Adoptive Transfer , Animals , Antigens, CD , Antigens, Differentiation/immunology , Blood/immunology , CTLA-4 Antigen , Cell Differentiation , Cell Division , Chickens , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...