Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Curr Hematol Malig Rep ; 11(3): 165-74, 2016 06.
Article in English | MEDLINE | ID: mdl-26939921

ABSTRACT

Acute lymphoblastic leukemia (ALL) among older adult patients presents significant clinical challenges. As opposed to pediatric populations, in whom long-term outcomes are markedly superior, those for adults remain grim. Nevertheless, younger adults with ALL have experienced a steady improvement in long-term survival in the last few decades. This is significantly different for older ALL patients, for whom long-term outcomes remain poor. Conventional chemotherapies are associated with sub-optimal outcomes and increased toxicity in this population. However, several emerging therapies, including antibody-drug conjugates, bi-specific engagers, and chimeric antigen receptor (CAR) T cells, have demonstrated much promise and are either incorporated into the existing therapeutic paradigms or being actively investigated to improve outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell- and Tissue-Based Therapy/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Antibodies, Bispecific/genetics , Antibodies, Bispecific/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Protein Kinase Inhibitors/therapeutic use , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Survival Rate
2.
Cancer J ; 20(2): 160-5, 2014.
Article in English | MEDLINE | ID: mdl-24667964

ABSTRACT

T-cell therapies using engineered T cells show great promise for cancer immunotherapy, as illustrated by the CD19 paradigm. Much of the excitement about this approach, and second-generation CARs in particular, is due to the dramatic clinical results recently reported by a few centers, especially in acute lymphoblastic leukemia, and the applicability of this approach, in principle, to a wide range of cancers. Extending the use of CAR therapies to cancers other than B-cell malignancies will require selective tumor targeting with minimal or acceptable "on-target, off-tumor" effects. The identification of new CAR target antigens is thus one of the next big challenges to address. Recognizing the paucity of currently available tumor-specific targets, we have developed broadly applicable approaches to enhance the tumor selectivity and safety of engineered T cells. Here, we review 2 promising concepts. One is to improve tumor targeting based on combinatorial antigen recognition. The other uses receptors that provide antigen-specific inhibition, which we named iCARs, to divert T cells from the normal tissues one wants to protect.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Antigen, T-Cell/therapeutic use , B-Lymphocytes/immunology , Epitopes , Humans , Neoplasms/genetics , Neoplasms/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/isolation & purification , T-Lymphocytes/immunology
3.
Sci Transl Med ; 5(215): 215ra172, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24337479

ABSTRACT

T cell therapies have demonstrated long-term efficacy and curative potential for the treatment of some cancers. However, their use is limited by damage to bystander tissues, as seen in graft-versus-host disease after donor lymphocyte infusion, or "on-target, off-tumor" toxicities incurred in some engineered T cell therapies. Nonspecific immunosuppression and irreversible T cell elimination are currently the only means to control such deleterious responses, but at the cost of abrogating therapeutic benefits or causing secondary complications. On the basis of the physiological paradigm of immune inhibitory receptors, we designed antigen-specific inhibitory chimeric antigen receptors (iCARs) to preemptively constrain T cell responses. We demonstrate that CTLA-4- or PD-1-based iCARs can selectively limit cytokine secretion, cytotoxicity, and proliferation induced through the endogenous T cell receptor or an activating chimeric receptor. The initial effect of the iCAR is temporary, thus enabling T cells to function upon a subsequent encounter with the antigen recognized by their activating receptor. iCARs thus provide a dynamic, self-regulating safety switch to prevent, rather than treat, the consequences of inadequate T cell specificity.


Subject(s)
CTLA-4 Antigen/chemistry , Immunotherapy/methods , Programmed Cell Death 1 Receptor/chemistry , Receptors, Antigen/chemistry , Animals , Cell Line , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/cytology , Fibroblasts/metabolism , Humans , Immunosuppression Therapy , Luminescence , Lymphocytes/cytology , Male , Mice , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Proteomics/methods , T-Lymphocytes/cytology
4.
Nat Biotechnol ; 31(10): 928-33, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23934177

ABSTRACT

Progress in adoptive T-cell therapy for cancer and infectious diseases is hampered by the lack of readily available, antigen-specific, human T lymphocytes. Pluripotent stem cells could provide an unlimited source of T lymphocytes, but the therapeutic potential of human pluripotent stem cell-derived lymphoid cells generated to date remains uncertain. Here we combine induced pluripotent stem cell (iPSC) and chimeric antigen receptor (CAR) technologies to generate human T cells targeted to CD19, an antigen expressed by malignant B cells, in tissue culture. These iPSC-derived, CAR-expressing T cells display a phenotype resembling that of innate γδ T cells. Similar to CAR-transduced, peripheral blood γδ T cells, the iPSC-derived T cells potently inhibit tumor growth in a xenograft model. This approach of generating therapeutic human T cells 'in the dish' may be useful for cancer immunotherapy and other medical applications.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/cytology , Animals , Antigens, CD19/metabolism , Cell Differentiation , Cell Proliferation , Chromium Radioisotopes , Cluster Analysis , Humans , Male , Mice , Phenotype , Protein Engineering , Receptors, Antigen , T-Lymphocytes/immunology
5.
Proc Natl Acad Sci U S A ; 110(29): 11851-6, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818587

ABSTRACT

Peptidylarginine deiminase 4 (PAD4) is a Ca(2+)-dependent enzyme that converts arginine and methylarginine residues to citrulline, with histone proteins being among its best-described substrates to date. However, the biological function of this posttranslational modification, either in histones or in nonhistone proteins, is poorly understood. Here, we show that PAD4 recognizes, binds, and citrullinates glycogen synthase kinase-3ß (GSK3ß), both in vitro and in vivo. Among other functions, GSK3ß is a key regulator of transcription factors involved in tumor progression, and its dysregulation has been associated with progression of human cancers. We demonstrate that silencing of PAD4 in breast cancer cells leads to a striking reduction of nuclear GSK3ß protein levels, increased TGF-ß signaling, induction of epithelial-to-mesenchymal transition, and production of more invasive tumors in xenograft assays. Moreover, in breast cancer patients, reduction of PAD4 and nuclear GSK3ß is associated with increased tumor invasiveness. We propose that PAD4-mediated citrullination of GSK3ß is a unique posttranslational modification that regulates its nuclear localization and thereby plays a critical role in maintaining an epithelial phenotype. We demonstrate a dynamic and previously unappreciated interplay between histone-modifying enzymes, citrullination of nonhistone proteins, and epithelial-to-mesenchymal transition.


Subject(s)
Citrulline/metabolism , Epithelial-Mesenchymal Transition/physiology , Glycogen Synthase Kinase 3/metabolism , Hydrolases/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Calcium Ionophores , Fluorescent Antibody Technique , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , MCF-7 Cells , Mass Spectrometry , Microscopy, Interference , Mutagenesis, Site-Directed , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
6.
J Clin Invest ; 120(12): 4410-4, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21084752

ABSTRACT

Recent genome-wide association studies have identified a genetic locus at human chromosome 8q24 as having minor alleles associated with lower levels of plasma triglyceride (TG) and LDL cholesterol (LDL-C), higher levels of HDL-C, as well as decreased risk for myocardial infarction. This locus contains only one annotated gene, tribbles homolog 1 (TRIB1), which has not previously been implicated in lipoprotein metabolism. Here we demonstrate a role for Trib1 as a regulator of lipoprotein metabolism in mice. Hepatic-specific overexpression of Trib1 reduced levels of plasma TG and cholesterol by reducing VLDL production; conversely, Trib1-knockout mice showed elevated levels of plasma TG and cholesterol due to increased VLDL production. Hepatic Trib1 expression was inversely associated with the expression of key lipogenic genes and measures of lipogenesis. Thus, we provide functional evidence for what we believe to be a novel gene regulating hepatic lipogenesis and VLDL production in mice that influences plasma lipids and risk for myocardial infarction in humans.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Lipid Metabolism/genetics , Liver/metabolism , Myocardial Infarction/genetics , Protein Serine-Threonine Kinases/genetics , APOBEC-1 Deaminase , Animals , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/metabolism , Lipogenesis/genetics , Lipoproteins, VLDL/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...