Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Epilepsia ; 63(8): 2037-2055, 2022 08.
Article in English | MEDLINE | ID: mdl-35560062

ABSTRACT

OBJECTIVE: Responsive neurostimulation is an effective therapy for patients with refractory mesial temporal lobe epilepsy. However, clinical outcomes are variable, few patients become seizure-free, and the optimal stimulation location is currently undefined. The aim of this study was to quantify responsive neurostimulation in the mesial temporal lobe, identify stimulation-dependent networks associated with seizure reduction, and determine if stimulation location or stimulation-dependent networks inform outcomes. METHODS: We modeled patient-specific volumes of tissue activated and created probabilistic stimulation maps of local regions of stimulation across a retrospective cohort of 22 patients with mesial temporal lobe epilepsy. We then mapped the network stimulation effects by seeding tractography from the volume of tissue activated with both patient-specific and normative diffusion-weighted imaging. We identified networks associated with seizure reduction across patients using the patient-specific tractography maps and then predicted seizure reduction across the cohort. RESULTS: Patient-specific stimulation-dependent connectivity was correlated with responsive neurostimulation effectiveness after cross-validation (p = .03); however, normative connectivity derived from healthy subjects was not (p = .44). Increased connectivity from the volume of tissue activated to the medial prefrontal cortex, cingulate cortex, and precuneus was associated with greater seizure reduction. SIGNIFICANCE: Overall, our results suggest that the therapeutic effect of responsive neurostimulation may be mediated by specific networks connected to the volume of tissue activated. In addition, patient-specific tractography was required to identify structural networks correlated with outcomes. It is therefore likely that altered connectivity in patients with epilepsy may be associated with the therapeutic effect and that utilizing patient-specific imaging could be important for future studies. The structural networks identified here may be utilized to target stimulation in the mesial temporal lobe and to improve seizure reduction for patients treated with responsive neurostimulation.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Epilepsy/therapy , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/therapy , Gyrus Cinguli , Humans , Magnetic Resonance Imaging , Retrospective Studies , Temporal Lobe
3.
Front Neurosci ; 15: 769872, 2021.
Article in English | MEDLINE | ID: mdl-34955721

ABSTRACT

Accurate anatomical localization of intracranial electrodes is important for identifying the seizure foci in patients with epilepsy and for interpreting effects from cognitive studies employing intracranial electroencephalography. Localization is typically performed by coregistering postimplant computed tomography (CT) with preoperative magnetic resonance imaging (MRI). Electrodes are then detected in the CT, and the corresponding brain region is identified using the MRI. Many existing software packages for electrode localization chain together separate preexisting programs or rely on command line instructions to perform the various localization steps, making them difficult to install and operate for a typical user. Further, many packages provide solutions for some, but not all, of the steps needed for confident localization. We have developed software, Locate electrodes Graphical User Interface (LeGUI), that consists of a single interface to perform all steps needed to localize both surface and depth/penetrating intracranial electrodes, including coregistration of the CT to MRI, normalization of the MRI to the Montreal Neurological Institute template, automated electrode detection for multiple types of electrodes, electrode spacing correction and projection to the brain surface, electrode labeling, and anatomical targeting. The software is written in MATLAB, core image processing is performed using the Statistical Parametric Mapping toolbox, and standalone executable binaries are available for Windows, Mac, and Linux platforms. LeGUI was tested and validated on 51 datasets from two universities. The total user and computational time required to process a single dataset was approximately 1 h. Automatic electrode detection correctly identified 4362 of 4695 surface and depth electrodes with only 71 false positives. Anatomical targeting was verified by comparing electrode locations from LeGUI to locations that were assigned by an experienced neuroanatomist. LeGUI showed a 94% match with the 482 neuroanatomist-assigned locations. LeGUI combines all the features needed for fast and accurate anatomical localization of intracranial electrodes into a single interface, making it a valuable tool for intracranial electrophysiology research.

4.
Brain Stimul ; 13(5): 1232-1244, 2020.
Article in English | MEDLINE | ID: mdl-32504827

ABSTRACT

BACKGROUND: Brain activity is constrained by and evolves over a network of structural and functional connections. Corticocortical evoked potentials (CCEPs) have been used to measure this connectivity and to discern brain areas involved in both brain function and disease. However, how varying stimulation parameters influences the measured CCEP across brain areas has not been well characterized. OBJECTIVE: To better understand the factors that influence the amplitude of the CCEPs as well as evoked gamma-band power (70-150 Hz) resulting from single-pulse stimulation via cortical surface and depth electrodes. METHODS: CCEPs from 4370 stimulation-response channel pairs were recorded across a range of stimulation parameters and brain regions in 11 patients undergoing long-term monitoring for epilepsy. A generalized mixed-effects model was used to model cortical response amplitudes from 5 to 100 ms post-stimulation. RESULTS: Stimulation levels <5.5 mA generated variable CCEPs with low amplitude and reduced spatial spread. Stimulation at ≥5.5 mA yielded a reliable and maximal CCEP across stimulation-response pairs over all regions. These findings were similar when examining the evoked gamma-band power. The amplitude of both measures was inversely correlated with distance. CCEPs and evoked gamma power were largest when measured in the hippocampus compared with other areas. Larger CCEP size and evoked gamma power were measured within the seizure onset zone compared with outside this zone. CONCLUSION: These results will help guide future stimulation protocols directed at quantifying network connectivity across cognitive and disease states.


Subject(s)
Cerebral Cortex/physiopathology , Deep Brain Stimulation/methods , Drug Resistant Epilepsy/diagnosis , Drug Resistant Epilepsy/physiopathology , Electrodes, Implanted , Gamma Rhythm/physiology , Adult , Brain Mapping/methods , Deep Brain Stimulation/instrumentation , Drug Resistant Epilepsy/therapy , Electroencephalography/methods , Evoked Potentials/physiology , Female , Humans , Male , Middle Aged , Random Allocation , Young Adult
5.
Cancer Discov ; 10(7): 998-1017, 2020 07.
Article in English | MEDLINE | ID: mdl-32349972

ABSTRACT

Loss-of-function mutations of EZH2, the enzymatic component of PRC2, have been associated with poor outcome and chemotherapy resistance in T-cell acute lymphoblastic leukemia (T-ALL). Using isogenic T-ALL cells, with and without CRISPR/Cas9-induced EZH2-inactivating mutations, we performed a cell-based synthetic lethal drug screen. EZH2-deficient cells exhibited increased sensitivity to structurally diverse inhibitors of CHK1, an interaction that could be validated genetically. Furthermore, small-molecule inhibition of CHK1 had efficacy in delaying tumor progression in isogenic EZH2-deficient, but not EZH2 wild-type, T-ALL cells in vivo, as well as in a primary cell model of PRC2-mutant ALL. Mechanistically, EZH2 deficiency resulted in a gene-expression signature of immature T-ALL cells, marked transcriptional upregulation of MYCN, increased replication stress, and enhanced dependency on CHK1 for cell survival. Finally, we demonstrate this phenotype is mediated through derepression of a distal PRC2-regulated MYCN enhancer. In conclusion, we highlight a novel and clinically exploitable pathway in high-risk EZH2-mutated T-ALL. SIGNIFICANCE: Loss-of-function mutations of PRC2 genes are associated with chemotherapy resistance in T-ALL, yet no specific therapy for this aggressive subtype is currently clinically available. Our work demonstrates that loss of EZH2 activity leads to MYCN-driven replication stress, resulting in increased sensitivity to CHK1 inhibition, a finding with immediate clinical relevance.This article is highlighted in the In This Issue feature, p. 890.


Subject(s)
Checkpoint Kinase 1/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Cell Proliferation , Humans , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
6.
Biomicrofluidics ; 13(1): 014107, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30867877

ABSTRACT

Renal disease is a global problem with unsustainable health-care costs. There currently exists a lack of accurate human renal disease models that take into account the complex microenvironment of these tissues. Here, we present a reusable microfluidic model of the human proximal tubule and glomerulus, which allows for the growth of renal epithelial cells in a variety of conditions that are representative of renal disease states including altered glomerular filtration rate, hyperglycemia, nephrolithiasis, and drug-induced nephrotoxicity (cisplatin and cyclosporine). Cells were exposed to these conditions under fluid flow or in traditional static cultures to determine the effects of a dynamic microenvironment on the pathogenesis of these renal disease states. The results indicate varying stress-related responses (α-smooth muscle actin (α-SMA) expression, alkaline phosphatase activity, fibronectin, and neutrophil gelatinase-associated lipocalin secretion) to each of these conditions when comparing cells that had been grown in static and dynamic conditions, potentially indicating more realistic and sensitive predictions of human responses and a requirement for a more complex "fit for purpose" model.

7.
Immunology ; 156(2): 130-135, 2019 02.
Article in English | MEDLINE | ID: mdl-30300924

ABSTRACT

T cells expressing chimeric antigen receptors (CARs) are a promising new cancer immunotherapy that has now reached the clinic. CARs are synthetic receptors that redirect T cells towards a tumour-associated antigen and activate them through various fused signalling regions, for example derived from CD3ζ, 4-1BB or CD28. Analysis of the optimal combination of CAR components including signalling domains is not yet comprehensive and may vary with the particular application. The C-terminus of the T-cell surface receptor CD6 is critical for its co-stimulatory effects and signals through two phospho-tyrosine motifs that bind to the intracellular adaptor proteins GADS and SLP-76. Addition of the C terminus of CD6 did not compromise CAR expression, showing it was a stable moiety that can be used independently of the native receptor. A third-generation CAR containing 4-1BB, CD3ζ and the C terminus of CD6 (4-1BBz-CD6) enhanced interferon-γ release and cytotoxicity when compared with the second-generation 4-1BB CD3ζ (4-1BBz) CAR. The CD6 C terminus is a valuable addition to potential components for modular design of CARs to improve effector function, particularly cytotoxicity.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Immunity, Cellular , Receptors, Chimeric Antigen/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line , Gene Expression , Mice , Phosphoproteins/genetics , Phosphoproteins/immunology , Protein Domains , Receptors, Chimeric Antigen/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
8.
Mol Ther ; 26(5): 1266-1276, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29661681

ABSTRACT

Engineered T cell therapies show considerable promise in the treatment of refractory malignancies. Given the ability of engineered T cells to engraft and persist for prolonged periods along with unpredicted toxicities, incorporation of a suicide gene to allow selective depletion after administration is desirable. Rapamycin is a safe and widely available immunosuppressive pharmaceutical that acts by heterodimerization of FKBP12 with the FRB fragment of mTOR. The apical caspase caspase 9 is activated by homodimerization through its CARD domain. We developed a rapamycin-induced caspase 9 suicide gene. First, we showed that caspase 9 could be activated by a two-protein format with replacement of the CARD domain with both FRB and FKBP12. We next identified an optimal compact single-protein rapamycin caspase 9 (rapaCasp9) by fusing both FRB and FKBP12 with the catalytic domain of caspase 9. Functionality of rapaCasp9 when co-expressed with a CD19 CAR was demonstrated in vitro and in vivo.


Subject(s)
Caspase 9/genetics , Gene Expression Regulation/drug effects , Gene Expression , Genes, Transgenic, Suicide , Sirolimus/pharmacology , Animals , Biomarkers , Caspase 9/chemistry , Caspase 9/metabolism , Cells, Cultured , Cytotoxicity, Immunologic , Genetic Vectors/genetics , Humans , Immunophenotyping , Mice , Protein Interaction Domains and Motifs , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Blood ; 131(7): 746-758, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29284597

ABSTRACT

B-cell maturation antigen (BCMA) is a promising therapeutic target for multiple myeloma (MM), but expression is variable, and early reports of BCMA targeting chimeric antigen receptors (CARs) suggest antigen downregulation at relapse. Dual-antigen targeting increases targetable tumor antigens and reduces the risk of antigen-negative disease escape. "A proliferation-inducing ligand" (APRIL) is a natural high-affinity ligand for BCMA and transmembrane activator and calcium-modulator and cyclophilin ligand (TACI). We quantified surface tumor expression of BCMA and TACI on primary MM cells (n = 50). All cases tested expressed BCMA, and 39 (78%) of them also expressed TACI. We engineered a third-generation APRIL-based CAR (ACAR), which killed targets expressing either BCMA or TACI (P < .01 and P < .05, respectively, cf. control, effector-to-target [E:T] ratio 16:1). We confirmed cytolysis at antigen levels similar to those on primary MM, at low E:T ratios (56.2% ± 3.9% killing of MM.1s at 48 h, E:T ratio 1:32; P < .01) and of primary MM cells (72.9% ± 12.2% killing at 3 days, E:T ratio 1:1; P < .05, n = 5). Demonstrating tumor control in the absence of BCMA, we maintained cytolysis of primary tumor expressing both BCMA and TACI in the presence of a BCMA-targeting antibody. Furthermore, using an intramedullary myeloma model, ACAR T cells caused regression of an established tumor within 2 days. Finally, in an in vivo model of tumor escape, there was complete ACAR-mediated tumor clearance of BCMA+TACI- and BCMA-TACI+ cells, and a single-chain variable fragment CAR targeting BCMA alone resulted in outgrowth of a BCMA-negative tumor. These results support the clinical potential of this approach.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B-Cell Maturation Antigen/metabolism , Multiple Myeloma/drug therapy , Receptors, Chimeric Antigen/therapeutic use , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Ligands , Mice , Molecular Targeted Therapy , Receptors, Chimeric Antigen/chemical synthesis , Receptors, Chimeric Antigen/chemistry , Transmembrane Activator and CAML Interactor Protein/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 13/chemistry
10.
Nat Med ; 23(12): 1416-1423, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29131157

ABSTRACT

Mature T cell cancers are typically aggressive, treatment resistant and associated with poor prognosis. Clinical application of immunotherapeutic approaches has been limited by a lack of target antigens that discriminate malignant from healthy (normal) T cells. Unlike B cell depletion, pan-T cell aplasia is prohibitively toxic. We report a new targeting strategy based on the mutually exclusive expression of T cell receptor ß-chain constant domains 1 and 2 (TRBC1 and TRBC2). We identify an antibody with unique TRBC1 specificity and use it to demonstrate that normal and virus-specific T cell populations contain both TRBC1+ and TRBC2+ compartments, whereas malignancies are restricted to only one. As proof of concept for anti-TRBC immunotherapy, we developed anti-TRBC1 chimeric antigen receptor (CAR) T cells, which recognized and killed normal and malignant TRBC1+, but not TRBC2+, T cells in vitro and in a disseminated mouse model of leukemia. Unlike nonselective approaches targeting the entire T cell population, TRBC-targeted immunotherapy could eradicate a T cell malignancy while preserving sufficient normal T cells to maintain cellular immunity.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy, Adoptive/methods , Leukemia, T-Cell/therapy , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Jurkat Cells , K562 Cells , Leukemia, T-Cell/immunology , Male , Mice , Molecular Targeted Therapy/methods , T-Lymphocytes/immunology
11.
Cytotherapy ; 18(8): 1002-1011, 2016 08.
Article in English | MEDLINE | ID: mdl-27378344

ABSTRACT

Novel cell therapies derived from human T lymphocytes are exhibiting enormous potential in early-phase clinical trials in patients with hematologic malignancies. Ex vivo modification of T cells is currently limited to a small number of centers with the required infrastructure and expertise. The process requires isolation, activation, transduction, expansion and cryopreservation steps. To simplify procedures and widen applicability for clinical therapies, automation of these procedures is being developed. The CliniMACS Prodigy (Miltenyi Biotec) has recently been adapted for lentiviral transduction of T cells and here we analyse the feasibility of a clinically compliant T-cell engineering process for the manufacture of T cells encoding chimeric antigen receptors (CAR) for CD19 (CAR19), a widely targeted antigen in B-cell malignancies. Using a closed, single-use tubing set we processed mononuclear cells from fresh or frozen leukapheresis harvests collected from healthy volunteer donors. Cells were phenotyped and subjected to automated processing and activation using TransAct, a polymeric nanomatrix activation reagent incorporating CD3/CD28-specific antibodies. Cells were then transduced and expanded in the CentriCult-Unit of the tubing set, under stabilized culture conditions with automated feeding and media exchange. The process was continuously monitored to determine kinetics of expansion, transduction efficiency and phenotype of the engineered cells in comparison with small-scale transductions run in parallel. We found that transduction efficiencies, phenotype and function of CAR19 T cells were comparable with existing procedures and overall T-cell yields sufficient for anticipated therapeutic dosing. The automation of closed-system T-cell engineering should improve dissemination of emerging immunotherapies and greatly widen applicability.


Subject(s)
Automation, Laboratory , Cell Engineering , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/metabolism , Automation, Laboratory/instrumentation , Automation, Laboratory/methods , B-Lymphocytes/immunology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Engineering/instrumentation , Cell Engineering/methods , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Computer-Aided Design , Humans , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/metabolism , Transduction, Genetic , Xenograft Model Antitumor Assays
12.
Cancer Res ; 75(18): 3853-64, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26183927

ABSTRACT

Adoptive immunotherapy using autologous T cells endowed with chimeric antigen receptors (CAR) has emerged as a powerful means of treating cancer. However, a limitation of this approach is that autologous CAR T cells must be generated on a custom-made basis. Here we show that electroporation of transcription activator-like effector nuclease (TALEN) mRNA allows highly efficient multiplex gene editing in primary human T cells. We use this TALEN-mediated editing approach to develop a process for the large-scale manufacturing of T cells deficient in expression of both their αß T-cell receptor (TCR) and CD52, a protein targeted by alemtuzumab, a chemotherapeutic agent. Functionally, T cells manufactured with this process do not mediate graft-versus-host reactions and are rendered resistant to destruction by alemtuzumab. These characteristics enable the administration of alemtuzumab concurrently or prior to engineered T cells, supporting their engraftment. Furthermore, endowing the TALEN-engineered cells with a CD19 CAR led to efficient destruction of CD19(+) tumor targets even in the presence of the chemotherapeutic agent. These results demonstrate the applicability of TALEN-mediated genome editing to a scalable process, which enables the manufacturing of third-party CAR T-cell immunotherapies against arbitrary targets. As such, CAR T-cell immunotherapies can therefore be used in an "off-the-shelf" manner akin to other biologic immunopharmaceuticals


Subject(s)
Gene Knockout Techniques , Immunotherapy, Adoptive , T-Lymphocytes/transplantation , Alemtuzumab , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/genetics , Antigens, CD19/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Base Sequence , CD52 Antigen , Cytotoxicity, Immunologic , Drug Resistance , Glycoproteins/deficiency , Glycoproteins/genetics , Graft vs Host Disease/prevention & control , Humans , Lymphocyte Activation , Lymphoma/therapy , Mice , Mice, Mutant Strains , Molecular Sequence Data , RNA, Messenger , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transfection , Xenograft Model Antitumor Assays
13.
Blood ; 124(8): 1277-87, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24970931

ABSTRACT

A compact marker/suicide gene that utilizes established clinical-grade reagents and pharmaceuticals would be of considerable practical utility to T-cell cancer gene therapy. Marker genes enable measurement of transduction and allow selection of transduced cells, whereas suicide genes allow selective deletion of administered T cells in the face of toxicity. We have created a highly compact marker/suicide gene for T cells combining target epitopes from both CD34 and CD20 antigens (RQR8). This construct allows selection with the clinically approved CliniMACS CD34 system (Miltenyi). Further, the construct binds the widely used pharmaceutical antibody rituximab, resulting in selective deletion of transgene-expressing cells. We have tested the functionality of RQR8 in vitro and in vivo as well as in combination with T-cell engineering components. We predict that RQR8 will make T-cell gene therapy both safer and cheaper.


Subject(s)
Antigens, CD20/immunology , Antigens, CD34/immunology , Genes, Transgenic, Suicide , Genetic Therapy/methods , Neoplasms/therapy , T-Lymphocytes/transplantation , Allografts , Animals , Antigens, CD20/genetics , Antigens, CD20/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Epitopes , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
Hum Gene Ther Methods ; 23(6): 376-86, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23186165

ABSTRACT

Use of adoptive T-cell therapy (ACT) is increasing; however, T-cell therapy can result in severe toxicity. Consequently, several suicide-gene strategies that allow selective destruction of the infused T cells have been described. We compared effectiveness of four such strategies in vitro in Epstein Barr virus (EBV)-cytotoxic T lymphocytes (CTLs). Herpes simplex virus thymidine kinase (HSV-TK), human inducible caspase 9 (iCasp9), mutant human thymidylate kinase (mTMPK), and human CD20 codon optimized genes were cloned in frame with 2A-truncated codon optimized CD34 (dCD34) in a retroviral vector. Codon-optimization considerably improved CD20 expression. EBV-CTLs could be efficiently transduced in all constructs, with transgene expression similar to the control vector containing dCD34 alone. Expression was maintained for prolonged cultures. Expression of the suicide genes was not associated with alterations in immunophenotype, proliferation, or function of CTLs. Activation of HSV-TK, iCasp9, and CD20 ultimately resulted in equally effective destruction of transduced T cells. However, while iCasp9 and CD20 effected immediate cell-death induction, HSV-TK-expressing T cells required 3 days of exposure to ganciclovir to reach full effect. mTMPK-transduced cells showed lower T-cell killing all time points. Our results suggest that the faster activity of iCasp9 might be advantageous in treating certain types of acutely life-threatening toxicity. Codon-optimized CD20 has potential as a suicide gene.


Subject(s)
Caspase 9/genetics , Genes, Transgenic, Suicide/genetics , T-Lymphocytes, Cytotoxic/metabolism , Thymidine Kinase/genetics , Antigens, CD20/genetics , Antigens, CD34/genetics , Ganciclovir/pharmacology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mutation , Simplexvirus/enzymology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
15.
JAMA ; 296(10): 1266-73, 2006 Sep 13.
Article in English | MEDLINE | ID: mdl-16968851

ABSTRACT

CONTEXT: Much of pediatric drug use is off-label because appropriate pediatric studies have not been conducted and the drugs have not been labeled by the US Food and Drug Administration (FDA) for use in children. In 1997, Congress authorized the FDA to grant extensions of marketing rights known as "pediatric exclusivity" if FDA-requested pediatric trials were conducted. As a result, there have been over 100 product labeling changes. The publication status of studies completed for pediatric exclusivity has not been evaluated. OBJECTIVE: To quantify the dissemination of results of studies conducted for pediatric exclusivity into the peer-review literature. DESIGN: Cohort study of all trials conducted for pediatric exclusivity between 1998 and 2004 as determined by MEDLINE and EMBASE searches through 2005, the subsequent labeling changes, and the publication of those studies in peer-reviewed journals. We categorized any labeling changes resulting from the studies as positive or negative for the drug under study. We then evaluated aspects of the studies and product label changes that were associated with subsequent publication in peer-reviewed medical journals. MAIN OUTCOME MEASURES: Publication of the trial data in peer-reviewed journals. RESULTS: Between 1998 and 2004, 253 studies were submitted to the FDA for pediatric exclusivity: 125 (50%) evaluated efficacy, 51 (20%) were multi-dose pharmacokinetic, 34 (13%) were single-dose pharmacokinetic, and 43 (17%) were safety studies. Labeling changes were positive for 127/253 (50%) of studies; only 113/253 (45%) were published. Efficacy studies and those with a positive labeling change were more likely to be published. CONCLUSIONS: The pediatric exclusivity program has been successful in encouraging drug studies in children. However, the dissemination of these results in the peer-reviewed literature is limited. Mechanisms to more widely disperse this information through publication warrant further evaluation.


Subject(s)
Clinical Trials as Topic , Pediatrics , Peer Review, Research , Pharmaceutical Preparations , Drug Labeling , United States , United States Food and Drug Administration
SELECTION OF CITATIONS
SEARCH DETAIL
...