Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 9(7)2021 07.
Article in English | MEDLINE | ID: mdl-34253637

ABSTRACT

BACKGROUND: Bispecific T cell engagers represent the majority of bispecific antibodies (BsAbs) entering the clinic to treat metastatic cancer. The ability to apply these agents safely and efficaciously in the clinic, particularly for solid tumors, has been challenging. Many preclinical studies have evaluated parameters related to the activity of T cell engaging BsAbs, but many questions remain. MAIN BODY: This study investigates the impact of affinity of T cell engaging BsAbs with regards to potency, efficacy, and induction of immunomodulatory receptors/ligands using HER-2/CD3 BsAbs as a model system. We show that an IgG BsAb can be as efficacious as a smaller BsAb format both in vitro and in vivo. We uncover a dichotomous relationship between tumor-associated antigen (TAA) affinity and CD3 affinity requirements for cells that express high versus low levels of TAA. HER-2 affinity directly correlated with the CD3 engager lysis potency of HER-2/CD3 BsAbs when HER-2 receptor numbers are high (~200 K/cell), while the CD3 affinity did not impact potency until its binding affinity was extremely low (<600 nM). When HER-2 receptor numbers were lower (~20 K/cell), both HER-2 and CD3 affinity impacted potency. The high affinity anti-HER-2/low CD3 affinity BsAb also demonstrated lower cytokine induction levels in vivo and a dosing paradigm atypical of extremely high potency T cell engaging BsAbs reaching peak efficacy at doses >3 mg/kg. This data confirms that low CD3 affinity provides an opportunity for improved safety and dosing for T cell engaging BsAbs. T cell redirection also led to upregulation of Programmed cell death 1 (PD-1) and 4-1BB, but not CTLA-4 on T cells, and to Programmed death-ligand 1 (PD-L1) upregulation on HER-2HI SKOV3 tumor cells, but not on HER-2LO OVCAR3 tumor cells. Using this information, we combined anti-PD-1 or anti-4-1BB monoclonal antibodies with the HER-2/CD3 BsAb in vivo and demonstrated significantly increased efficacy against HER-2HI SKOV3 tumors via both combinations. CONCLUSIONS: Overall, these studies provide an informational dive into the optimization process of CD3 engaging BsAbs for solid tumors indicating that a reduced affinity for CD3 may enable a better therapeutic index with a greater selectivity for the target tumor and a reduced cytokine release syndrome. These studies also provide an additional argument for combining T cell checkpoint inhibition and co-stimulation to achieve optimal efficacy.


Subject(s)
Antibodies, Bispecific/therapeutic use , T-Lymphocytes/immunology , Animals , Antibodies, Bispecific/pharmacology , Humans , Mice
2.
PLoS One ; 16(2): e0245917, 2021.
Article in English | MEDLINE | ID: mdl-33596227

ABSTRACT

Pre-clinical murine models are critical for translating drug candidates from the bench to the bedside. There is interest in better understanding how anti-human CD3 therapy works based on recent longitudinal studies of short-term administration. Although several models have been created in this pursuit, each have their own advantages and disadvantages in Type-1 diabetes. In this study, we report a murine genetic knock-in model which expresses both a murine and a humanized-CD3ε-exon, rendering it sensitive to manipulation with anti-human CD3. These huCD3εHET mice are viable and display no gross abnormalities. Specifically, thymocyte development and T cell peripheral homeostasis is unaffected. We tested immune functionality of these mice by immunizing them with T cell-dependent antigens and no differences in antibody titers compared to wild type mice were recorded. Finally, we performed a graft-vs-host disease model that is driven by effector T cell responses and observed a wasting disease upon transfer of huCD3εHET T cells. Our results show a viable humanized CD3 murine model that develops normally, is functionally engaged by anti-human CD3 and can instruct on pre-clinical tests of anti-human CD3 antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , CD3 Complex/genetics , CD3 Complex/immunology , Gene Knock-In Techniques , Animals , Humans , Mice , Mice, Inbred C57BL , Phenotype , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymocytes/cytology , Thymocytes/immunology
3.
J Clin Aesthet Dermatol ; 9(7): 13-5, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27672407

ABSTRACT

In recent years, a large number of therapeutic monoclonal antibodies have come to market to treat a variety of conditions including patients with immune-mediated chronic inflammation. Distinguishing the relative clinical efficacy and safety profiles of one monoclonal antibody relative to another can be difficult and complex due to different clinical designs and paucity of head-to-head comparator studies. One distinguishing feature in interpreting clinical trial data by dermatologists may begin by determining whether a monoclonal antibody is fully human or humanized, which can be discerned by the generic name of the drug. Herein, this commentary highlights the distinctions and similarities of fully human and humanized monoclonal antibodies in their nomenclature, engineering, and clinical profiles. While there are a number of differences between these types of monoclonal antibodies, current evidence indicates that this designation does not impart any measurable impact on overall clinical efficacy and safety profiles of a given drug. Based on molecular insights provided in this commentary, it is clear that each monoclonal antibody, irrespective of being fully human or humanized, should be individually assessed for its clinical impact regarding safety and efficacy. Going beyond the type of generic name ascribed to a monoclonal antibody will be an ever-increasing theme for dermatologists as more therapeutic monoclonal antibodies emerge to potentially treat a wider scope of diseases with cutaneous manifestations.

4.
Biol Psychiatry ; 80(1): 12-22, 2016 07 01.
Article in English | MEDLINE | ID: mdl-26831917

ABSTRACT

BACKGROUND: The mechanisms underlying stress-induced inflammation that contribute to major depressive disorder are unknown. We examine the role of the adenosine triphosphate (ATP)/purinergic type 2X7 receptor (P2X7R) pathway and the NLRP3 (nucleotide-binding, leucine-rich repeat, pyrin domain containing 3) inflammasome in interleukin (IL)-1ß and depressive behavioral responses to stress. METHODS: The influence of acute restraint stress on extracellular ATP, glutamate, IL-1ß, and tumor necrosis factor alpha in hippocampus was determined by microdialysis, and the influence of acute restraint stress on the NLRP3 inflammasome was determined by western blot analysis. The influence of P2X7R antagonist administration on IL-1ß and tumor necrosis factor alpha and on anxiety and depressive behaviors was determined in the chronic unpredictable stress rodent model. The role of the NLRP3 inflammasome was determined by analysis of Nlrp3 null mice. RESULTS: Acute restraint stress rapidly increased extracellular ATP, an endogenous agonist of P2X7R; the inflammatory cytokine IL-1ß; and the active form of the NLRP3 inflammasome in the hippocampus. Administration of a P2X7R antagonist completely blocked the release of IL-1ß and tumor necrosis factor alpha, another stress-induced cytokine, and activated NLRP3. Moreover, P2X7R antagonist administration reversed the anhedonic and anxiety behaviors caused by chronic unpredictable stress exposure, and deletion of the Nlrp3 gene rendered mice resistant to development of depressive behaviors caused by chronic unpredictable stress. CONCLUSIONS: These findings demonstrate that psychological "stress" is sensed by the innate immune system in the brain via the ATP/P2X7R-NLRP3 inflammasome cascade, and they identify novel therapeutic targets for the treatment of stress-related mood disorders and comorbid illnesses.


Subject(s)
Adenosine Triphosphate/metabolism , Anxiety/metabolism , Behavior, Animal/physiology , Depression/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Purinergic P2Y2/metabolism , Stress, Psychological/metabolism , Tumor Necrosis Factor-alpha/metabolism , Anhedonia/physiology , Animals , Anxiety/etiology , Anxiety/immunology , Depression/etiology , Depression/immunology , Disease Models, Animal , Male , Mice , Mice, Knockout , Purinergic P2Y Receptor Agonists/metabolism , Purinergic P2Y Receptor Antagonists/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications , Stress, Psychological/immunology
5.
MAbs ; 7(3): 470-82, 2015.
Article in English | MEDLINE | ID: mdl-25774965

ABSTRACT

A myriad of innovative bispecific antibody (BsAb) platforms have been reported. Most require significant protein engineering to be viable from a development and manufacturing perspective. Single-chain variable fragments (scFvs) and diabodies that consist only of antibody variable domains have been used as building blocks for making BsAbs for decades. The drawback with Fv-only moieties is that they lack the native-like interactions with CH1/CL domains that make antibody Fab regions stable and soluble. Here, we utilize a redesigned Fab interface to explore 2 novel Fab-based BsAbs platforms. The redesigned Fab interface designs limit heavy and light chain mixing when 2 Fabs are co-expressed simultaneously, thus allowing the use of 2 different Fabs within a BsAb construct without the requirement of one or more scFvs. We describe the stability and activity of a HER2×HER2 IgG-Fab BsAb, and compare its biophysical and activity properties with those of an IgG-scFv that utilizes the variable domains of the same parental antibodies. We also generated an EGFR × CD3 tandem Fab protein with a similar format to a tandem scFv (otherwise known as a bispecific T cell engager or BiTE). We show that the Fab-based BsAbs have superior biophysical properties compared to the scFv-based BsAbs. Additionally, the Fab-based BsAbs do not simply recapitulate the activity of their scFv counterparts, but are shown to possess unique biological activity.


Subject(s)
Antibodies, Bispecific , Recombinant Fusion Proteins , Single-Chain Antibodies , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Cell Line, Tumor , Humans , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
6.
J Immunol ; 175(4): 2286-92, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16081797

ABSTRACT

Genetic disruption of death receptor 6 (DR6) results in enhanced CD4+ T cell expansion, Th2 differentiation, and humoral responses after stimulation. However, the in vivo consequences of DR6 targeting (DR6-/-) during the initiation and progression of inflammatory autoimmune disease are unclear. Using a myelin oligodendrocyte glycoprotein (MOG(35-55))-induced model of experimental autoimmune encephalomyelitis, DR6-/- mice were found to be highly resistant to both the onset and the progression of CNS disease compared with wild-type (WT) littermates. DR6-/- mice exhibited fewer inflammatory foci along with minimal demyelination and perivascular cuffing of inflammatory cells. Consistent with these observations, mononuclear cell infiltration, including CD4+ T cells and macrophages, in the spinal cord of DR6-/- mice was dramatically reduced. Furthermore, CD4+ T cells from DR6-/- mice exhibited profoundly reduced cell surface expression of VLA-4 before and after stimulation. Compared with WT mice, DR6-/- mice exhibited significantly increased autoantigen-induced T cell proliferative responses along with greater numbers of IL-4-producing and similar or slightly higher numbers of IFN-gamma-producing CD4+ T cells. DR6-/- CD4+ T cells secreted higher levels of the Th2 cytokine, IL-4, and similar levels of the Th1 cytokine, IFN-gamma, compared with WT cells. Taken together, our data demonstrate that DR6 plays an important role in regulating leukocyte infiltration and function in the induction and progression of experimental autoimmune encephalomyelitis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Glycoproteins/administration & dosage , Glycoproteins/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/transplantation , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/pathology , Cell Movement/genetics , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Down-Regulation/genetics , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epitopes, T-Lymphocyte/immunology , Female , Immunity, Innate/genetics , Immunization, Passive , Integrin alpha4beta1/antagonists & inhibitors , Integrin alpha4beta1/biosynthesis , Interleukin-4/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Count , Mice , Mice, Knockout , Molecular Sequence Data , Myelin-Oligodendrocyte Glycoprotein , Receptors, Tumor Necrosis Factor/physiology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Up-Regulation/genetics , Up-Regulation/immunology
7.
Immunology ; 110(2): 225-33, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14511236

ABSTRACT

Fas ligand (FasL)-induced lung inflammation has recently been suggested to play an important role in the pathogenesis of acute respiratory disease syndrome (ARDS). In order to further explore this connection, we established a FasL-induced murine model of pulmonary inflammation. Instillation of recombinant FasL (rFasL) into the lung induced neutrophil infiltration and increased pulmonary permeability, as evidenced by increased total protein in the airspace; both occur in patients with ARDS. These effects were accompanied with a rapid induction of proinflammatory mediators: cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) and the chemokines macrophage inflammatory protein-2 (MIP-2) and KC. Pretreatment with a FasL antagonist, a decoy receptor 3 analogue (DcR3 analogue), reduced neutrophil infiltration into the airspace and resulted in a highly significant reduction in the levels of GM-CSF, MIP-2 and KC in bronchoalveolar lavage (BAL) fluid. We postulate that rFasL may be responsible for induction of proinflammatory chemokines and cytokines in the lung, which in turn attract neutrophil infiltration into the airspace. This proinflammatory process and the associated pulmonary permeability may, in part, explain the association of FasL with severe pulmonary inflammation, such as ARDS, and shed new light on FasL and its role in lung injury.


Subject(s)
Membrane Glycoproteins/immunology , Membrane Glycoproteins/therapeutic use , Receptors, Cell Surface/therapeutic use , Respiratory Distress Syndrome/immunology , Animals , Chemokine CXCL2 , Disease Models, Animal , Fas Ligand Protein , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Ligands , Lipopolysaccharides/immunology , Macrophages, Alveolar/immunology , Membrane Glycoproteins/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Monokines/metabolism , Neutrophil Infiltration/immunology , Peritonitis/immunology , Receptors, Tumor Necrosis Factor , Receptors, Tumor Necrosis Factor, Member 6b , Respiratory Distress Syndrome/prevention & control
8.
Int Immunol ; 15(7): 861-70, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12807825

ABSTRACT

LIGHT, a newly identified member of the tumor necrosis factor (TNF) family, is expressed on activated T lymphocytes. To evaluate how LIGHT contributes to T cell functions, we generated LIGHT-deficient (LIGHT(-/-)) mice using gene targeting. Disruption of LIGHT significantly reduced CD8(+) T cell-cycle progression, leading to reduced proliferation to anti-CD3, anti-CD3/anti-CD28 or allogeneic stimulation, whereas proliferation of CD4(+) T cells remained unchanged. In contrast to the observed proliferative defects, isolated CD8(+) T cells from LIGHT(-/-) mice displayed normal cytotoxic effector function development when compared to wild-type CD8(+) T cells. Underlying a potential mechanism of reduced CD8(+) T cell proliferation, LIGHT(-/-) CD8(+) T cells displayed reduced surface levels of CD25 and a diminished ability to proliferate in response to exogenous IL-2. Furthermore, addition of IL-12 to LIGHT(-/-) CD8(+) T cell cultures could not ameliorate this proliferative defect. These results reveal a potential mechanism of action for LIGHT as a positive regulator of CD8(+) T cell expansion, but not lytic effector function development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Membrane Proteins/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis , Genetic Vectors , Interleukin-12/pharmacology , Interleukin-2/pharmacology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interleukin-2/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14 , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
J Exp Med ; 197(1): 51-62, 2003 Jan 06.
Article in English | MEDLINE | ID: mdl-12515813

ABSTRACT

Targeted disruption of death receptor (DR)6 results in enhanced CD4(+) T cell expansion and T helper cell type 2 differentiation after stimulation. Similar to T cells, DR6 is expressed on resting B cells but is down-regulated upon activation. We examined DR6(-/-) B cell responses both in vitro and in vivo. In vitro, DR6(-/-) B cells undergo increased proliferation in response to anti-immunoglobulin M, anti-CD40, and lipopolysaccharide. This hyperproliferative response was due, at least in part, to both increased cell division and reduced cell apoptosis when compared with wild-type B cells. Consistent with these observations, increased nuclear levels and activity of nuclear factor kappaB transcription factor, c-Rel, and elevated Bcl-x(l) expression were observed in DR6(-/-) B cells upon stimulation. In addition, DR6(-/-) B cells exhibited higher surface levels of CD86 upon activation and were more effective as antigen-presenting cells in an allogeneic T cell proliferation response. DR6(-/-) mice exhibited enhanced germinal center formation and increased titers of immunoglobulins to T-dependent as well as T-independent type I and II antigens. This is the first demonstration of a regulatory role of DR6 in the activation and function of B cells.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Lymphocyte Activation , Receptors, Tumor Necrosis Factor/metabolism , Animals , Antigen Presentation , Antigens, CD/metabolism , Apoptosis , B-Lymphocytes/cytology , B7-2 Antigen , Cell Division , Cell Survival , Cells, Cultured , Down-Regulation , Flow Cytometry , Gene Deletion , Immunohistochemistry , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Mitogens/immunology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , T-Lymphocytes/immunology , Up-Regulation , bcl-X Protein
10.
Clin Cancer Res ; 8(10): 3270-5, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12374698

ABSTRACT

The selective estrogen receptor modulator arzoxifene and the rexinoid LG 100268 were active not only as single agents for prevention and treatment of breast cancer in the rat model that uses nitrosomethylurea as the carcinogen but also showed striking synergy, both preventively and therapeutically, in a series of six experiments with a total of 465 rats. Mechanistic studies in cell culture reported here suggest that enhancement of stromal-epithelial interactions may contribute to this synergy. The possible clinical use of the combination of arzoxifene and LG 100268 for prevention of breast cancer in women at high risk, for treatment of women in the adjuvant setting, or for treatment of end-stage disease should now be considered.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Mammary Neoplasms, Experimental/prevention & control , Nicotinic Acids/therapeutic use , Piperidines/therapeutic use , Selective Estrogen Receptor Modulators/therapeutic use , Tetrahydronaphthalenes/therapeutic use , Thiophenes/therapeutic use , Animals , Cells, Cultured , Drug Synergism , Drug Therapy, Combination , Female , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea , Neoplasm Invasiveness , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Rats , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism
11.
J Immunol ; 169(7): 3993-8, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12244201

ABSTRACT

DR6 is a recently identified member of the TNFR family. In a previous study, we have shown that DR6 KO mice have enhanced CD4(+) T cell proliferation and Th2 cytokine production. Acute graft-vs-host disease (GVHD) results from the activation and expansion of alloreactive donor T cells following bone marrow transplantation. In this article, we demonstrate that the transfer of donor T cells from DR6 KO mice into allogeneic recipient mice in a parent into an F(1) model of acute GVHD results in a more rapid onset of GVHD with increased severity. Recipients of DR6 KO T cells exhibit earlier systemic symptoms of GVHD, more rapid weight loss, earlier histopathological organ damage in the thymus, spleen, and intestines, and earlier mortality. The rapid onset of GVHD in these mice may be attributable to the enhanced activation and expansion of DR6 KO CD4(+) and CD8(+) T cells. Our findings support the hypothesis that DR6 serves as an important regulatory molecule in T cell immune responses. The identification and use of DR6 ligands and/or agonistic Abs to DR6 may represent useful therapeutics in the treatment of T cell-mediated diseases such as GVHD.


Subject(s)
Adoptive Transfer , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , T-Lymphocyte Subsets/transplantation , Acute Disease , Adoptive Transfer/methods , Animals , B-Lymphocytes/pathology , Cell Division/genetics , Cell Division/immunology , Cytotoxicity, Immunologic/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/mortality , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Isoantigens/immunology , Lymphocyte Activation/genetics , Lymphocyte Culture Test, Mixed , Lymphopenia/genetics , Lymphopenia/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Receptors, Tumor Necrosis Factor/administration & dosage , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/pathology , Tumor Cells, Cultured , Weight Loss/genetics , Weight Loss/immunology
12.
J Neurochem ; 80(6): 1039-48, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11953454

ABSTRACT

Activated caspase-3 is considered an important enzyme in the cell death pathway. To study the specific role of caspase-3 activation in neuronal cells, we generated a stable tetracycline-regulated SK-N-MC neuroblastoma cell line, which expressed a highly efficient self-activating chimeric caspase-3, consisting of the caspase-1 prodomain fused to the caspase-3 catalytic domain. Under expression-inducing conditions, we observed a time-dependent increase of processed caspase-3 by immunostaining for the active form of the enzyme, intracellular caspase-3 enzyme activity, as well as poly(ADP-ribose) polymerase (PARP) cleavage. Induced expression of the caspase fusion protein showed predominantly caspase-3 activity without any apoptotic morphological changes. In contrast, staurosporine treatment of the same cells resulted in activation of multiple caspases and profound apoptotic morphology. Our work provides evidence that auto-activation of caspase-3 can be efficiently achieved with a longer prodomain and that neuronal cell apoptosis may require another caspase or activation of multiple caspase enzymes.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Neuroblastoma/metabolism , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspase 3 , Caspases/genetics , Cell Survival/drug effects , Doxycycline/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Neuroblastoma/pathology , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Staurosporine/pharmacology , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL