Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Pharmacol Res Perspect ; 3(6): e00199, 2015 Dec.
Article En | MEDLINE | ID: mdl-27022472

Hu714MuXHu is a recombinant chimeric murine-human monoclonal antibody directed against interleukin-15 (IL-15), a proinflammatory cytokine associated with memory CD8+ and natural killer (NK) T-cell activation and implicated in the pathogenesis of inflammatory diseases. A pharmacokinetic-pharmacodynamic (PK/PD) model was developed to describe the NK cell count reduction in cynomolgus monkeys after treatment with Hu714MuXHu. Cynomolgus monkeys were dosed with Hu714MuXHu in three studies: as a single dose at 0.1 or 1 mg·kg(-1) i.v.; weekly for 5 weeks at 0, 30, 60, or 150 mg·kg(-1) i.v. or 150 mg·kg(-1) s.c.; weekly for 13 weeks at 0, 5, 30, or 150 mg·kg(-1) s.c. Serum Hu714MuXHu concentration-time data were analyzed using noncompartmental analysis and the PK/NK cell count relationship was assessed via simultaneous PK/PD modeling. Hu714MuXHu PK was approximately dose-proportional between 0.1-150 mg·kg(-1) for i.v. and 5-150 mg·kg(-1) for s.c. administration with an elimination half-life of 12.7-18 days. Hu714MuXHu administration resulted in rapid and marked reductions in NK cell counts after the first dose which recovered fully after the serum Hu714MuXHu concentrations approached 0.1 µg·mL(-1) (assay limit of quantification). PK/PD modeled Hu714MuXHu effects on NK cells had an EC 50 of 0.09 µg·mL(-1). In summary, weekly i.v. or s.c. doses with Hu714MuXHu for up to 3 months in cynomolgus monkeys demonstrated linear PK and significant NK cell count reduction, which was described using PK/PD modeling. This approach may be used to guide investigative product dose selections for inflammatory diseases where NK cell count alterations are quantifiable.

2.
J Immunol ; 191(11): 5551-8, 2013 Dec 01.
Article En | MEDLINE | ID: mdl-24184554

IL-15 is a proinflammatory cytokine that plays an important role in the development and activation of NK cells and is a potential target for inflammatory disease therapy. Studies conducted in IL-15- and IL-15R knockout mice identified IL-15 as an important cytokine for NK cell homeostasis. Consistent with this information derived from genetically modified mice, we demonstrated that neutralizing IL-15 with a mouse anti-mouse IL-15 mAb (M96) depletes C57BL/6 mouse NK cells. An mAb directed against macaque IL-15 (Hu714MuXHu) was manufactured and demonstrated to block IL-15-induced activation of nonhuman primate (NHP) NK cells in vitro. Neutralization of macaque IL-15 by parenteral administration of Hu714MuXHu reduces (>95%) circulating NK cell counts in NHPs. A blocking mAb directed against human IL-15 (huIL-15; AMG 714) was manufactured. Unexpectedly, when human subjects were treated with the blocking anti-IL-15 Ab AMG 714 in clinical trials, no reductions in circulating NK cell counts were observed despite achieving significantly higher exposures than the levels of Hu714MuXHu needed to cause NK cell count reductions in NHPs in vivo. Both AMG 714 and Hu714MuXHu are able to block huIL-15 activity in a human T cell blast proliferation and IFN-γ production assay. Both Abs block huIL-15-mediated Stat5 activation and CD69 expression in human NK cells. Collectively, these results demonstrate that NK cell homeostasis is obligatorily dependent upon IL-15 in both mice and NHPs, but that IL-15 is dispensable for maintenance of circulating human NK cells.


Homeostasis , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Animals , Antibodies, Blocking/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Clinical Trials as Topic , Homeostasis/drug effects , Humans , Interferon-gamma/metabolism , Interleukin-15/genetics , Interleukin-15/immunology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Macaca , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT5 Transcription Factor/metabolism , Transcriptional Activation/drug effects
3.
J Clin Invest ; 120(6): 2131-43, 2010 Jun.
Article En | MEDLINE | ID: mdl-20440074

Enteropathy-associated T cell lymphoma is a severe complication of celiac disease (CD). One mechanism suggested to underlie its development is chronic exposure of intraepithelial lymphocytes (IELs) to potent antiapoptotic signals initiated by IL-15, a cytokine overexpressed in the enterocytes of individuals with CD. However, the signaling pathway by which IL-15 transmits these antiapoptotic signals has not been firmly established. Here we show that the survival signals delivered by IL-15 to freshly isolated human IELs and to human IEL cell lines derived from CD patients with type II refractory CD (RCDII) - a clinicopathological entity considered an intermediary step between CD and enteropathy-associated T cell lymphoma - depend on the antiapoptotic factors Bcl-2 and/or Bcl-xL. The signals also required IL-15Rbeta, Jak3, and STAT5, but were independent of PI3K, ERK, and STAT3. Consistent with these data, IELs from patients with active CD and RCDII contained increased amounts of Bcl-xL, phospho-Jak3, and phospho-STAT5. Furthermore, incubation of patient duodenal biopsies with a fully humanized human IL-15-specific Ab effectively blocked Jak3 and STAT5 phosphorylation. In addition, treatment with this Ab induced IEL apoptosis and wiped out the massive IEL accumulation in mice overexpressing human IL-15 in their gut epithelium. Together, our results delineate the IL-15-driven survival pathway in human IELs and demonstrate that IL-15 and its downstream effectors are meaningful therapeutic targets in RCDII.


Apoptosis/immunology , Celiac Disease/immunology , Inflammation/immunology , Interleukin-15/immunology , Lymphocytes/immunology , Adult , Apoptosis/drug effects , Celiac Disease/complications , Celiac Disease/metabolism , Cytokines/immunology , Cytokines/metabolism , Cytokines/pharmacology , Enterocytes/immunology , Enterocytes/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Interleukin-15/metabolism , Interleukin-15/pharmacology , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestines/immunology , Janus Kinase 3/immunology , Janus Kinase 3/metabolism , Leukemia/complications , Leukemia/immunology , Leukemia/metabolism , Lymphocytes/metabolism , Phosphorylation , Protein Binding/immunology , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology
4.
J Immunol ; 180(5): 2912-21, 2008 Mar 01.
Article En | MEDLINE | ID: mdl-18292513

The role of costimulation has previously been confined to the very early stages of the CD8+ T cell response. In this study, we demonstrate the requirement for CD27 costimulation during the later phase, but not programming of the primary CD8+ T cell response to influenza virus and reveal a novel mechanism of action for CD27 costimulation. CD27 signals, during the later phase of the primary CD8+ T cell response, prevent apoptosis of Ag-specific CD8+ T cells. Blocking CD27L (CD70) on days 6 and 8 after infection reduces the number of NP(366-374)-specific CD8+ T cells, increases their sensitivity to CD95/Fas-mediated apoptosis, and up-regulates FasL on CD4+ T cells. This reduction of NP(366-374)-specific CD8+ T cells requires the presence of CD4+ T cells and Fas signaling. Lack of CD27 signals also decreases the quality of memory CD8+ T cell responses. Memory CD8+ T cells, which express surface CD27 similar to naive cells, however, do not require CD27 costimulation during a secondary response. Thus, CD27 acts indirectly to regulate primary Ag-specific CD8+ T cell responses by preventing apoptosis of CD8+ T cells during the later phase of the primary response and is required for optimal quality of memory cells, but is not required during normally primed secondary CD8+ T cell responses.


Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/physiology , fas Receptor/physiology , Adoptive Transfer , Animals , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Apoptosis/genetics , CD27 Ligand/antagonists & inhibitors , CD27 Ligand/immunology , CD27 Ligand/metabolism , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Immunization, Secondary , Immunologic Memory/genetics , Influenza A virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Signal Transduction/genetics , Time Factors , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Viral Core Proteins/administration & dosage , Viral Core Proteins/immunology , fas Receptor/biosynthesis , fas Receptor/deficiency , fas Receptor/genetics
5.
J Exp Med ; 195(12): 1515-22, 2002 Jun 17.
Article En | MEDLINE | ID: mdl-12070279

Both naive and memory T cells undergo antigen-independent proliferation after transfer into a T cell-depleted environment (acute homeostatic proliferation), whereas only memory T cells slowly divide in a full T cell compartment (basal proliferation). We show, first, that naive and memory CD8+ T cells have different cytokine requirements for acute homeostatic proliferation. Interleukin (IL)-7 receptor(R)alpha-mediated signals were obligatory for proliferation of naive T cells in lymphopenic hosts, whereas IL-15 did not influence their division. Memory T cells, on the other hand, could use either IL-7Ralpha- or IL-15-mediated signals for acute homeostatic proliferation: their proliferation was delayed when either IL-7Ralpha was blocked or IL-15 removed, but only when both signals were absent was proliferation ablated. Second, the cytokine requirements for basal and acute homeostatic proliferation of CD8+ memory T cells differ, as basal division of memory T cells was blocked completely in IL-15-deficient hosts. These data suggest a possible mechanism for the dearth of memory CD8+ T cells in IL-15- and IL-15Ralpha-deficient mice is their impaired basal proliferation. Our results show that naive and memory T lymphocytes differ in their cytokine dependence for acute homeostatic proliferation and that memory T lymphocytes have distinct requirements for proliferation in full versus empty compartments.


CD8-Positive T-Lymphocytes/immunology , Cell Division/physiology , Homeostasis/physiology , Immunologic Memory/physiology , Interleukin-15/physiology , Interleukin-7/physiology , Animals , Base Sequence , CD8-Positive T-Lymphocytes/cytology , DNA Primers , Female , Flow Cytometry , Mice , Mice, Inbred C57BL
...