Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Am J Respir Crit Care Med ; 209(4): 362-373, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38113442

ABSTRACT

Despite progress in elucidation of disease mechanisms, identification of risk factors, biomarker discovery, and the approval of two medications to slow lung function decline in idiopathic pulmonary fibrosis and one medication to slow lung function decline in progressive pulmonary fibrosis, pulmonary fibrosis remains a disease with a high morbidity and mortality. In recognition of the need to catalyze ongoing advances and collaboration in the field of pulmonary fibrosis, the NHLBI, the Three Lakes Foundation, and the Pulmonary Fibrosis Foundation hosted the Pulmonary Fibrosis Stakeholder Summit on November 8-9, 2022. This workshop was held virtually and was organized into three topic areas: 1) novel models and research tools to better study pulmonary fibrosis and uncover new therapies, 2) early disease risk factors and methods to improve diagnosis, and 3) innovative approaches toward clinical trial design for pulmonary fibrosis. In this workshop report, we summarize the content of the presentations and discussions, enumerating research opportunities for advancing our understanding of the pathogenesis, treatment, and outcomes of pulmonary fibrosis.


Subject(s)
Biomedical Research , Idiopathic Pulmonary Fibrosis , United States , Humans , National Heart, Lung, and Blood Institute (U.S.) , Lakes , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/therapy , Risk Factors
2.
Int J Mol Sci ; 22(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070369

ABSTRACT

Folate receptor beta (FRß) is a folate binding receptor expressed on myeloid lineage hematopoietic cells. FRß is commonly expressed at high levels on malignant blasts in patients with acute myeloid leukemia (AML), as well as on M2 polarized tumor-associated macrophages (TAMs) in the tumor microenvironment of many solid tumors. Therefore, FRß is a potential target for both direct and indirect cancer therapy. We demonstrate that FRß is expressed in both AML cell lines and patient-derived AML samples and that a high-affinity monoclonal antibody against FRß (m909) has the ability to cause dose- and expression-dependent ADCC against these cells in vitro. Importantly, we find that administration of m909 has a significant impact on tumor growth in a humanized mouse model of AML. Surprisingly, m909 functions in vivo with and without the infusion of human NK cells as mediators of ADCC, suggesting potential involvement of mouse macrophages as effector cells. We also found that TAMs from primary ovarian ascites samples expressed appreciable levels of FRß and that m909 has the ability to cause ADCC in these samples. These results indicate that the targeting of FRß using m909 has the potential to limit the outgrowth of AML in vitro and in vivo. Additionally, m909 causes cytotoxicity to TAMs in the tumor microenvironment of ovarian cancer warranting further investigation of m909 and its derivatives as therapeutic agents in patients with FRß-expressing cancers.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Folate Receptor 2 , Immunotherapy , Leukemia, Myeloid, Acute , Neoplasm Proteins , Ovarian Neoplasms , Animals , CHO Cells , Cricetulus , Female , Folate Receptor 2/antagonists & inhibitors , Folate Receptor 2/immunology , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , THP-1 Cells , Xenograft Model Antitumor Assays
3.
J Immunol ; 193(1): 185-97, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24899507

ABSTRACT

Inhibitors of Bruton's tyrosine kinase (BTK) possess much promise for the treatment of oncologic and autoimmune indications. However, our current knowledge of the role of BTK in immune competence has been gathered in the context of genetic inactivation of btk in both mice and man. Using the novel BTK inhibitor PF-303, we model the clinical phenotype of BTK inhibition by systematically examining the impact of PF-303 on the mature immune system in mice. We implicate BTK in tonic BCR signaling, demonstrate dependence of the T3 B cell subset and IgM surface expression on BTK activity, and find that B1 cells survive and function independently of BTK. Although BTK inhibition does not impact humoral memory survival, Ag-driven clonal expansion of memory B cells and Ab-secreting cell generation are inhibited. These data define the role of BTK in the mature immune system and mechanistically predict the clinical phenotype of chronic BTK inhibition.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunity, Humoral/physiology , Immunologic Memory/physiology , Models, Immunological , Protein-Tyrosine Kinases/immunology , Agammaglobulinaemia Tyrosine Kinase , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Humans , Immunoglobulin M/genetics , Immunoglobulin M/immunology , Mice , Mice, Inbred CBA , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics
4.
J Immunol ; 191(9): 4540-50, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24068666

ABSTRACT

Autoantibody production and immune complex deposition within the kidney promote renal disease in patients with lupus nephritis. Thus, therapeutics that inhibit these pathways may be efficacious in the treatment of systemic lupus erythematosus. Bruton's tyrosine kinase (BTK) is a critical signaling component of both BCR and FcR signaling. We sought to assess the efficacy of inhibiting BTK in the development of lupus-like disease, and in this article describe (R)-5-amino-1-(1-cyanopiperidin-3-yl)-3-(4-[2,4-difluorophenoxy]phenyl)-1H-pyrazole-4-carboxamide (PF-06250112), a novel highly selective and potent BTK inhibitor. We demonstrate in vitro that PF-06250112 inhibits both BCR-mediated signaling and proliferation, as well as FcR-mediated activation. To assess the therapeutic impact of BTK inhibition, we treated aged NZBxW_F1 mice with PF-06250112 and demonstrate that PF-06250112 significantly limits the spontaneous accumulation of splenic germinal center B cells and plasma cells. Correspondingly, anti-dsDNA and autoantibody levels were reduced in a dose-dependent manner. Moreover, administration of PF-06250112 prevented the development of proteinuria and improved glomerular pathology scores in all treatment groups. Strikingly, this therapeutic effect could occur with only a modest reduction observed in anti-dsDNA titers, implying a critical role for BTK signaling in disease pathogenesis beyond inhibition of autoantibody production. We subsequently demonstrate that PF-06250112 prevents proteinuria in an FcR-dependent, Ab-mediated model of glomerulonephritis. Importantly, these results highlight that BTK inhibition potently limits the development of glomerulonephritis by impacting both cell- and effector molecule-mediated pathways. These data provide support for evaluating the efficacy of BTK inhibition in systemic lupus erythematosus patients.


Subject(s)
B-Lymphocytes/immunology , Glomerulonephritis/immunology , Lupus Erythematosus, Systemic/immunology , Piperidines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/therapeutic use , Agammaglobulinaemia Tyrosine Kinase , Animals , B-Lymphocytes/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Germinal Center/cytology , Glomerulonephritis/metabolism , Glomerulonephritis/prevention & control , Kidney/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/prevention & control , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NZB , Piperidines/pharmacology , Plasma Cells/drug effects , Plasma Cells/immunology , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Receptors, Fc , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
5.
J Immunol ; 188(4): 1656-67, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22231702

ABSTRACT

MRL/MpJ-Fas(lpr/lpr)/J (MRL(lpr)) mice develop lupus-like disease manifestations in an IL-21-dependent manner. IL-21 is a pleiotropic cytokine that can influence the activation, differentiation, and expansion of B and T cell effector subsets. Notably, autoreactive CD4(+) T and B cells spontaneously accumulate in MRL(lpr) mice and mediate disease pathogenesis. We sought to identify the particular lymphocyte effector subsets regulated by IL-21 in the context of systemic autoimmunity and, thus, generated MRL(lpr) mice deficient in IL-21R (MRL(lpr).IL-21R(-/-)). Lymphadenopathy and splenomegaly, which are characteristic traits of the MRL(lpr) model were significantly reduced in the absence of IL-21R, suggesting that immune activation was likewise decreased. Indeed, spontaneous germinal center formation and plasma cell accumulation were absent in IL-21R-deficient MRL(lpr) mice. Correspondingly, we observed a significant reduction in autoantibody titers. Activated CD4(+) CD44(+) CD62L(lo) T cells also failed to accumulate, and CD4(+) Th cell differentiation was impaired, as evidenced by a significant reduction in CD4(+) T cells that produced the pronephritogenic cytokine IFN-γ. T extrafollicular helper cells are a recently described subset of activated CD4(+) T cells that function as the primary inducers of autoantibody production in MRL(lpr) mice. Importantly, we demonstrated that T extrafollicular helper cells are dependent on IL-21R for their generation. Together, our data highlighted the novel observation that IL-21 is a critical regulator of multiple pathogenic B and T cell effector subsets in MRL(lpr) mice.


Subject(s)
Autoimmunity , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukins/immunology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Receptors, Interleukin-21/immunology , Animals , Autoantibodies/genetics , Autoantibodies/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Interferon-gamma/biosynthesis , Lymphatic Diseases/genetics , Lymphatic Diseases/immunology , Lymphatic Diseases/pathology , Mice , Mice, Inbred MRL lpr , Mice, Knockout , Receptors, Interleukin-21/deficiency , Receptors, Interleukin-21/genetics , Skin/immunology , Skin/pathology , Splenomegaly/genetics , Splenomegaly/immunology , Splenomegaly/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology
6.
J Immunol ; 186(2): 667-74, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21169545

ABSTRACT

Development of long-term humoral immunity, characterized by the formation of long-lived plasma cells (PCs) in the bone marrow and memory B cells, is a critical component of protective immunity to pathogens, and as such it is the major goal of vaccination. However, the mechanisms involved in the generation of long-term humoral immunity remain poorly understood. In this study, we used IL-21R-deficient (IL-21R.KO) mice to examine the role of the IL-21 pathway in the development of the B cell memory response. Primary IgG serum Ab responses to the T cell-dependent Ag 4-hydroxy-3-nitrophenylacetyl (NP) hapten conjugated to chicken γ globulin were delayed in IL-21R.KO mice, but reached normal titers within 3 to 4 wk of immunization. IL-21R.KO mice formed germinal centers and generated normal numbers of PCs in their bone marrow. Additionally, memory B cell formation was similar in wild-type and IL-21R.KO mice. However, NP-specific memory B cells and PCs failed to expand following secondary immunization of IL-21R.KO mice, and consequently, secondary IgG Ab responses to NP hapten conjugated to chicken γ globulin were significantly impaired. These results identify the IL-21 pathway as a critical component of the memory B cell response.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Immunologic Memory , Receptors, Interleukin-21/physiology , Animals , Antigens, Surface/biosynthesis , Apoptosis Regulatory Proteins/biosynthesis , B-Lymphocyte Subsets/cytology , Cell Differentiation/genetics , Chickens/immunology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Haptens/administration & dosage , Haptens/immunology , Immunization, Secondary , Immunologic Memory/genetics , Immunologic Memory/immunology , Leukocyte Common Antigens/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitrophenols/administration & dosage , Nitrophenols/immunology , Phenylacetates/administration & dosage , Phenylacetates/immunology , Programmed Cell Death 1 Receptor , Receptors, CXCR5/biosynthesis , Receptors, Interleukin-21/deficiency , Receptors, Interleukin-21/genetics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , gamma-Globulins/administration & dosage , gamma-Globulins/immunology
7.
Cell Immunol ; 278(1-2): 103-12, 2012.
Article in English | MEDLINE | ID: mdl-23121982

ABSTRACT

The infiltration of neutrophils and monocytes is a prominent feature of inflammatory diseases including human rheumatoid arthritis. Understanding how neutrophil recruitment is regulated during pathogenesis is crucial for developing anti-inflammatory therapies. We optimized the K/B×N serum-induced mouse arthritis model to study neutrophil trafficking dynamics in vivo using two-photon microscopy. Arthritogenic serum was injected subcutaneously into one hind footpad to induce a local arthritis with robust neutrophil recruitment. Using this approach, we showed that the depletion of monocytes with clodronate liposomes impaired neutrophil recruitment specifically at the transendothelial migration step. The depletion of CCR2(+) monocytes with the monoclonal antibody MC-21 reproduced these effects, implicating CCR2(+) monocytes as key regulators of neutrophil extravasation during arthritis initiation. However, monocyte depletion did not prevent neutrophil extravasation in response to bacterial challenge. These findings suggest that anti-inflammatory therapies targeting monocytes may act in part through antagonizing neutrophil extravasation at sites of aseptic inflammation.


Subject(s)
Arthritis, Experimental/immunology , Monocytes/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Animals , Antibodies, Monoclonal/pharmacology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Clodronic Acid/pharmacology , Female , Humans , Injections, Subcutaneous , Liposomes , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Monocytes/drug effects , Monocytes/pathology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/pathology , Receptors, CCR2/biosynthesis
8.
Arthritis Rheum ; 63(10): 2939-48, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21618207

ABSTRACT

OBJECTIVE: Interleukin-22 (IL-22) is a mediator in antimicrobial responses and inflammatory autoimmune diseases. Although IL-22 and its receptor, IL-22R, have been identified in the synovium of rheumatoid arthritis patients, the source of IL-22 and its contribution to disease pathogenicity remain to be established. This study was undertaken to investigate the regulation of IL-22 by Th17 cells in vitro and to evaluate the potential for IL-22 depletion in an experimental arthritis model using mice deficient in the IL-1 receptor antagonist (IL-1Ra-/-). METHODS: Naive murine T cells were cultured under conditions leading to polarization of the cells into subsets of Th1, Th2, induced Treg, and Th17. Cytokines were measured in the culture supernatants, and the cells were analyzed by fluorescence-activated cell sorting. Tissue samples from the inflamed ankle synovium of IL-1Ra-/- mice were isolated, and messenger RNA levels of marker genes were quantified. IL-1Ra-/- mice were treated with neutralizing anti-IL-22 antibodies. Synovial cells were isolated from the inflamed tissue and sorted into fractions for analysis of cytokine production. RESULTS: In vitro tests showed that Th17 cells produced high levels of IL-22 after stimulation with IL-1 or IL-23. Interestingly, a synergistic increase in the production of IL-22 was observed after combining IL-1 and IL-23. In vivo, IL-1Ra-/- mice displayed a progressive erosive arthritis, characterized by up-regulation of IL-17 in mildly and severely inflamed tissue, whereas the levels of IL-22 and IL-22R were increased only in severely inflamed synovia. Anti-IL-22 treatment of IL-1Ra-/- mice significantly reduced the inflammation and bone erosion. Analysis of isolated single cells from the inflamed synovia revealed that IL-22 was mainly produced by IL-17-expressing T cells. CONCLUSION: These findings suggest that IL-22 plays an important role in IL-1-driven chronic joint destruction.


Subject(s)
Arthritis, Experimental/immunology , Bone and Bones/metabolism , Interleukin-1/metabolism , Interleukins/metabolism , Synovial Membrane/metabolism , Th17 Cells/immunology , Animals , Arthritis, Experimental/metabolism , Bone and Bones/pathology , Cell Differentiation , Inflammation/metabolism , Interleukin-23/metabolism , Joints/metabolism , Joints/pathology , Mice , Th17 Cells/metabolism , Interleukin-22
9.
J Immunol ; 185(9): 5531-8, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20870942

ABSTRACT

IL-22 is made by a unique set of innate and adaptive immune cells, including the recently identified noncytolytic NK, lymphoid tissue-inducer, Th17, and Th22 cells. The direct effects of IL-22 are restricted to nonhematopoietic cells, its receptor expressed on the surface of only epithelial cells and some fibroblasts in various organs, including parenchymal tissue of the gut, lung, skin, and liver. Despite this cellular restriction on IL-22 activity, we demonstrate that IL-22 induces effects on systemic biochemical, cellular, and physiological parameters. By utilizing adenoviral-mediated delivery of IL-22 and systemic administration of IL-22 protein, we observed that IL-22 modulates factors involved in coagulation, including fibrinogen levels and platelet numbers, and cellular constituents of blood, such as neutrophil and RBC counts. Furthermore, we observed that IL-22 induces thymic atrophy, body weight loss, and renal proximal tubule metabolic activity. These cellular and physiological parameters are indicative of a systemic inflammatory state. We observed that IL-22 induces biochemical changes in the liver including induction of fibrinogen, CXCL1, and serum amyloid A that likely contribute to the reported cellular and physiological effects of IL-22. Based on these findings, we propose that downstream of its expression and impact in local tissue inflammation, circulating IL-22 can further induce changes in systemic physiology that is indicative of an acute-phase response.


Subject(s)
Acute-Phase Reaction/immunology , Acute-Phase Reaction/physiopathology , Interleukins/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Interleukin-22
10.
J Clin Invest ; 118(2): 597-607, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202747

ABSTRACT

Psoriasis is a chronic skin disease resulting from the dysregulated interplay between keratinocytes and infiltrating immune cells. We report on a psoriasis-like disease model, which is induced by the transfer of CD4(+)CD45RB(hi)CD25(-) cells to pathogen-free scid/scid mice. Psoriasis-like lesions had elevated levels of antimicrobial peptide and proinflammatory cytokine mRNA. Also, similar to psoriasis, disease progression in this model was dependent on the p40 common to IL-12 and IL-23. To investigate the role of IL-22, a Th17 cytokine, in disease progression, mice were treated with IL-22-neutralizing antibodies. Neutralization of IL-22 prevented the development of disease, reducing acanthosis (thickening of the skin), inflammatory infiltrates, and expression of Th17 cytokines. Direct administration of IL-22 into the skin of normal mice induced both antimicrobial peptide and proinflammatory cytokine gene expression. Our data suggest that IL-22, which acts on keratinocytes and other nonhematopoietic cells, is required for development of the autoreactive Th17 cell-dependent disease in this model of skin inflammation. We propose that IL-22 antagonism might be a promising therapy for the treatment of human psoriasis.


Subject(s)
Dermatitis/immunology , Interleukins/physiology , Psoriasis/immunology , Skin/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Blocking/pharmacology , Cytokines/blood , Disease Models, Animal , Female , Gene Expression/drug effects , Interleukin-12/pharmacology , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-17/metabolism , Interleukins/antagonists & inhibitors , Interleukins/pharmacology , Lipopolysaccharides/immunology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/immunology , Interleukin-22
11.
Rheumatology (Oxford) ; 50(6): 1033-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21258049

ABSTRACT

OBJECTIVES: To characterize the in vitro binding and effector function properties of CD20-directed small modular immunopharmaceutical (SMIP) 2LM20-4, and to compare its in vivo B-cell depletion activity with the mutated 2LM20-4 P331S [no in vitro complement-dependent cytotoxicity (CDC)] and rituximab in cynomolgus monkeys. METHODS: Direct binding is examined in flow cytometry, confocal microscopy, scatchard and lipid raft assays. Effector function assays include CDC and Fc-mediated cellular toxicity. In the 6-month-long in vivo B-cell depletion study, single i.v. dosages of 1 or 10 mg/kg of anti-CD20 proteins were administered to monkeys and B-cell counts were monitored in peripheral blood, bone marrow and lymph nodes. RESULTS: 2LM20-4 has lower saturation binding to human primary B cells and recruits fewer CD20 molecules into lipid rafts compared with rituximab; however, it induces higher in vitro CDC. In competitive binding, 2LM20-4 only partially displaces rituximab, suggesting that it binds to a fraction of CD20 molecules within certain locations of the plasma membrane as compared with rituximab. In monkeys, 2LM20-4 had more sustained B-cell depletion activity than rituximab in peripheral blood and had significantly more profound and sustained activity than 2LM20-4 P331S and rituximab in the lymph nodes. CONCLUSIONS: SMIP 2LM20-4, which binds to a fraction of CD20 molecules as compared with rituximab, has more potent in vitro CDC, and more potent and sustained B-cell depletion activity in cynomolgus monkeys. Our work has considerable clinical relevance since it provides novel insights related to the emerging B-cell depletion therapies in autoimmune diseases.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD20/drug effects , Antigens, CD20/immunology , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Binding Sites , Cells, Cultured , Disease Models, Animal , Humans , Immunologic Factors/pharmacology , In Vitro Techniques , Linear Models , Macaca fascicularis , Random Allocation , Rituximab , Sensitivity and Specificity
12.
Bioorg Med Chem Lett ; 21(21): 6258-63, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21958547

ABSTRACT

Imidazo[1,5-a]quinoxalines were synthesized that function as irreversible Bruton's tyrosine kinase (BTK) inhibitors. The syntheses and SAR of this series of compounds are presented as well as the X-ray crystal structure of the lead compound 36 in complex with a gate-keeper variant of ITK enzyme. The lead compound showed good in vivo efficacy in preclinical RA models.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinoxalines/pharmacology , Agammaglobulinaemia Tyrosine Kinase , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Quinoxalines/chemistry
13.
Arthritis Rheum ; 62(8): 2283-93, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20506481

ABSTRACT

OBJECTIVE: All gamma-chain cytokines signal through JAK-3 and JAK-1 acting in tandem. We undertook this study to determine whether the JAK-3 selective inhibitor WYE-151650 would be sufficient to disrupt cytokine signaling and to ameliorate autoimmune disease pathology without inhibiting other pathways mediated by JAK-1, JAK-2, and Tyk-2. METHODS: JAK-3 kinase selective compounds were characterized by kinase assay and JAK-3-dependent (interleukin-2 [IL-2]) and -independent (IL-6, granulocyte-macrophage colony-stimulating factor [GM-CSF]) cell-based assays measuring proliferation or STAT phosphorylation. In vivo, off-target signaling was measured by IL-22- and erythropoietin (EPO)-mediated models, while on-target signaling was measured by IL-2-mediated signaling. Efficacy of JAK-3 inhibitors was determined using delayed-type hypersensitivity (DTH) and collagen-induced arthritis (CIA) models in mice. RESULTS: In vitro, WYE-151650 potently suppressed IL-2-induced STAT-5 phosphorylation and cell proliferation, while exhibiting 10-29-fold less activity against JAK-3-independent IL-6- or GM-CSF-induced STAT phosphorylation. Ex vivo, WYE-151650 suppressed IL-2-induced STAT phosphorylation, but not IL-6-induced STAT phosphorylation, as measured in whole blood. In vivo, WYE-151650 inhibited JAK-3-mediated IL-2-induced interferon-gamma production and decreased the natural killer cell population in mice, while not affecting IL-22-induced serum amyloid A production or EPO-induced reticulocytosis. WYE-151650 was efficacious in mouse DTH and CIA models. CONCLUSION: In vitro, ex vivo, and in vivo assays demonstrate that WYE-151650 is efficacious in mouse CIA despite JAK-3 selectivity. These data question the need to broadly inhibit JAK-1-, JAK-2-, or Tyk-2-dependent cytokine pathways for efficacy.


Subject(s)
Arthritis, Experimental/drug therapy , Janus Kinase 3/antagonists & inhibitors , Analysis of Variance , Animals , Arthritis, Experimental/metabolism , Blotting, Western , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Signal Transduction/drug effects
14.
J Exp Med ; 197(10): 1297-302, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12743172

ABSTRACT

Pathogenic mechanisms relevant to rheumatoid arthritis occur in the mouse model of collagen-induced arthritis (CIA). Cytosolic phospholipase A2alpha (cPLA2alpha) releases arachidonic acid from cell membranes to initiate the production of prostaglandins and leukotrienes. These inflammatory mediators have been implicated in the development of CIA. To test the hypothesis that cPLA2alpha plays a key role in the development of CIA, we backcrossed cPLA2alpha-deficient mice on the DBA/1LacJ background that is susceptible to CIA. The disease severity scores and the incidence of disease were markedly reduced in cPLA2alpha-deficient mice compared with wild-type littermates. At completion of the study, >90% of the wild-type mice had developed disease whereas none of the cPLA2alpha-deficient mice had more than one digit inflamed. Furthermore, visual disease scores correlated with severity of disease determined histologically. Pannus formation, articular fibrillation, and ankylosis were all dramatically reduced in the cPLA2alpha-deficient mice. Although the disease scores differed significantly between cPLA2alpha mutant and wild-type mice, anti-collagen antibody levels were similar in the wild-type mice and mutant littermates. These data demonstrate the critical role of cPLA2alpha in the pathogenesis of CIA.


Subject(s)
Arthritis/etiology , Collagen/immunology , Cytosol/enzymology , Phospholipases A/physiology , 5-Lipoxygenase-Activating Proteins , Animals , Arthritis/prevention & control , Carrier Proteins/physiology , Cyclooxygenase 2 , Group IV Phospholipases A2 , Immunization , Immunoglobulin G/blood , Immunoglobulin G/classification , Isoenzymes/physiology , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Phospholipases A/deficiency , Prostaglandin-Endoperoxide Synthases/physiology , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP4 Subtype
15.
Cell Immunol ; 266(1): 14-23, 2010.
Article in English | MEDLINE | ID: mdl-20851384

ABSTRACT

Delayed-type hypersensitivity (DTH) is classically defined as inflammation involving activated Th1 cells and cytokine production. DTH paw swelling, along with the cytokines IL-2, IFNγ, MCP-1 and TNFα, were inhibited in Balb/c mice by cyclosporine A (CsA). Surprisingly, the DTH response in the B6D2F1 mice was unaffected by CsA, despite a decrease in TNFα and IFNγ levels. IL-2 levels, however, were not decreased. To determine if the IL-2 production in the B6D2F1 strain is occurring through CD28-mediated costimulation, both CsA and CTLA-4Ig were administered. Paw swelling and IL-2 levels were decreased, indicating a role for costimulation. Co-administration of temsirolimus and CsA also reduced DTH and IL-2 levels in B6D2F1 mice, demonstrating involvement of the mTORC1 pathway. These results indicate that the cell activation pathways responsible for DTH differ with mouse strain. It is important to understand these differences in order to accurately interpret the results using potential therapeutic agents.


Subject(s)
Autoimmunity/immunology , Cyclosporine/pharmacology , Hypersensitivity, Delayed/immunology , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Interleukin-2/immunology , Abatacept , Animals , Antibodies/immunology , Antibodies/pharmacology , Autoimmunity/drug effects , CD28 Antigens/metabolism , Cell Movement/drug effects , Cell Movement/immunology , Cyclosporine/pharmacokinetics , Cytokines/metabolism , Erythrocytes/immunology , Female , Foot/pathology , Hypersensitivity, Delayed/pathology , Immunoconjugates/pharmacology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Multiprotein Complexes , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/metabolism , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Sheep , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Species Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases , Vaccination
16.
Sci Transl Med ; 12(567)2020 10 28.
Article in English | MEDLINE | ID: mdl-33115948

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a lethal disease with an average life expectancy of 3 to 5 years. IPF is characterized by progressive stiffening of the lung parenchyma due to excessive deposition of collagen, leading to gradual failure of gas exchange. Although two therapeutic agents have been approved from the FDA for IPF, they only slow disease progression with little impact on outcome. To develop a more effective therapy, we have exploited the fact that collagen-producing myofibroblasts express a membrane-spanning protein, fibroblast activation protein (FAP), that exhibits limited if any expression on other cell types. Because collagen-producing myofibroblasts are only found in fibrotic tissues, solid tumors, and healing wounds, FAP constitutes an excellent marker for targeted delivery of drugs to tissues undergoing pathologic fibrosis. We demonstrate here that a low-molecular weight FAP ligand can be used to deliver imaging and therapeutic agents selectively to FAP-expressing cells. Because induction of collagen synthesis is associated with phosphatidylinositol 3-kinase (PI3K) activation, we designed a FAP-targeted PI3K inhibitor that selectively targets FAP-expressing human IPF lung fibroblasts and potently inhibited collagen synthesis. Moreover, we showed that administration of the inhibitor in a mouse model of IPF inhibited PI3K activation in fibrotic lungs, suppressed production of hydroxyproline (major building block of collagen), reduced collagen deposition, and increased mouse survival. Collectively, these studies suggest that a FAP-targeted PI3K inhibitor might be promising for treating IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Phosphatidylinositol 3-Kinases , Animals , Fibroblasts , Idiopathic Pulmonary Fibrosis/drug therapy , Lung , Mice , Models, Theoretical , TOR Serine-Threonine Kinases
17.
EMBO Mol Med ; 12(8): e12034, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32597014

ABSTRACT

Fibrotic diseases cause organ failure that lead to ~45% of all deaths in the United States. Activated macrophages stimulate fibrosis by secreting cytokines that induce fibroblasts to synthesize collagen and extracellular matrix proteins. Although suppression of macrophage-derived cytokine production can halt progression of fibrosis, therapeutic agents that prevent release of these cytokines (e.g., TLR7 agonists) have proven too toxic to administer systemically. Based on the expression of folate receptor ß solely on activated myeloid cells, we have created a folate-targeted TLR7 agonist (FA-TLR7-54) that selectively accumulates in profibrotic macrophages and suppresses fibrosis-inducing cytokine production. We demonstrate that FA-TLR7-54 reprograms M2-like fibrosis-inducing macrophages into fibrosis-suppressing macrophages, resulting in dramatic declines in profibrotic cytokine release, hydroxyproline biosynthesis, and collagen deposition, with concomitant increases in alveolar airspaces. Although nontargeted TLR7-54 is lethal at fibrosis-suppressing doses, FA-TLR7-54 halts fibrosis without evidence of toxicity. Taken together, FA-TLR7-54 is shown to constitute a novel and potent approach for treating fibrosis without causing dose-limiting systemic toxicities.


Subject(s)
Bleomycin , Pulmonary Fibrosis , Animals , Fibroblasts , Macrophages , Macrophages, Alveolar , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy
18.
Arthritis Res Ther ; 21(1): 143, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31174578

ABSTRACT

OBJECTIVES: Most therapies for autoimmune and inflammatory diseases either neutralize or suppress production of inflammatory cytokines produced by activated macrophages (e.g., TNFα, IL-1, IL-6, IL-17, GM-CSF). However, no approved therapies directly target this activated subset of macrophages. METHODS: First, we undertook to examine whether the folate receptor beta (FR-ß) positive subpopulation of macrophages, which marks the inflammatory subset in animal models of rheumatoid arthritis, might constitute the prominent population of macrophages in inflamed lesions in humans. Next, we utilized anti-FR-ß monoclonal antibodies capable of mediating antibody-dependent cell cytotoxicity (ADCC) to treat animal models of rheumatoid arthritis and peritonitis. RESULTS: Human tissue samples of rheumatoid arthritis, Crohn's disease, ulcerative colitis, idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, chronic obstructive pulmonary disease, systemic lupus erythematosus, psoriasis, and scleroderma are all characterized by dramatic accumulation of macrophages that express FR-ß, a protein not expressed on resting macrophages or any other healthy tissues. A monoclonal antibody to FR-ß accumulates specifically in inflamed lesions of murine inflammatory disease models and successfully treats such models of rheumatoid arthritis and peritonitis. More importantly, elimination of FR-ß-positive macrophages upon treatment with an anti-FR-ß monoclonal antibody promotes the departure of other immune cells, including T cells, B cells, neutrophils, and dendritic cells from the inflamed lesions. CONCLUSIONS: These data suggest that specific elimination of FR-ß-expressing macrophages may constitute a highly specific therapy for multiple autoimmune and inflammatory diseases and that a recently developed human anti-human FR-ß monoclonal antibody (m909) might contribute to suppression of this subpopulation of macrophages.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/immunology , Folate Receptor 2/immunology , Immunity, Cellular , Macrophage Activation , Macrophages/metabolism , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Macrophages/immunology , Macrophages/pathology , Mice
19.
J Med Chem ; 51(12): 3388-413, 2008 Jun 26.
Article in English | MEDLINE | ID: mdl-18498150

ABSTRACT

The optimization of a class of indole cPLA 2 alpha inhibitors is described herein. The importance of the substituent at C3 and the substitution pattern of the phenylmethane sulfonamide region are highlighted. Optimization of these regions led to the discovery of 111 (efipladib) and 121 (WAY-196025), which are shown to be potent, selective inhibitors of cPLA 2 alpha in a variety of isolated enzyme assays, cell based assays, and rat and human whole blood assays. The binding of these compounds has been further examined using isothermal titration calorimetry. Finally, these compounds have shown efficacy when dosed orally in multiple acute and chronic prostaglandin and leukotriene dependent in vivo models.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Benzoates/chemical synthesis , Group IV Phospholipases A2/antagonists & inhibitors , Sulfonamides/chemical synthesis , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/drug therapy , Benzoates/chemistry , Benzoates/pharmacology , Biological Availability , Bronchoconstriction/drug effects , Calorimetry , Carrageenan , Cell Line , Cyclooxygenase 2 Inhibitors/chemical synthesis , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Edema/chemically induced , Edema/drug therapy , Humans , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Male , Mice , Protein Binding , Rats , Rats, Sprague-Dawley , Sheep , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
20.
Bioorg Med Chem ; 16(3): 1345-58, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17998165

ABSTRACT

The synthesis and structure-activity relationship of a series of benzenesulfonamide indole inhibitors of cPLA(2)alpha are described. Substitution of the benzenesulfonamide led to analogues with 50-fold improvement in potency versus the unsubstituted benzenesulfonamide lead compound. Rat pharmacokinetics in a minimal formulation was used to prioritize compounds, leading to the discovery of a potent inhibitor of cPLA(2)alpha with oral efficacy in models of rat carrageenan paw edema and Ascaris suum airway challenge in naturally sensitized sheep.


Subject(s)
Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/metabolism , Indoles/pharmacology , Sulfonamides/chemistry , Administration, Oral , Animals , Ascariasis/drug therapy , Ascariasis/parasitology , Ascaris suum/physiology , Calorimetry , Humans , Indoles/chemistry , Indoles/therapeutic use , Molecular Structure , Rats , Sheep , Structure-Activity Relationship , Temperature , Benzenesulfonamides
SELECTION OF CITATIONS
SEARCH DETAIL