Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Clin Drug Investig ; 41(1): 65-76, 2021 Jan.
Article En | MEDLINE | ID: mdl-33331980

BACKGROUND AND OBJECTIVES: Matrix metalloproteinases (MMPs) are proteases with different biological and pathological activities, and many have been linked to several diseases. Targeting individual MMPs may offer a safer therapeutic potential for several diseases. We assessed the safety, tolerability, and pharmacokinetics of FP-025, a novel, highly selective oral matrix metalloproteinase-12 inhibitor, in healthy subjects. METHODS: Two randomized, double-blind, placebo-controlled studies were conducted. Study I was a first-in-man study, evaluating eight single ascending doses (SADs) (50-800 mg) in two formulations: i.e., neat FP-025 in capsule (API-in-Capsule) and in an amorphous solid dispersion (ASD-in-Capsule) formulation. In Study II, three multiple ascending doses (MADs) (100, 200, and 400 mg, twice daily) of FP-025 (ASD-in-Capsule) were administered for 8 days, including a food-effect evaluation. RESULTS: Ninety-six subjects were dosed. Both formulations were well tolerated with one adverse event (AE) reported in the 800 mg API-in-Capsule SAD group and seven AEs throughout the MAD groups. The exposure to FP-025 was low with the API-in-Capsule formulation; it increased dose-dependently with the ASD-in-Capsule formulation, with which exposure to FP-025 increased in a greater-than-dose-proportional manner at lower doses (≤ 100 mg) but less proportionally at higher doses. The elimination half-life (t1/2) was between 6 (Study I) and 8 h (Study II). Accumulation of FP-025 was approximately 1.7-fold in the MAD study. Food intake delayed the rate of absorption, but without effect in the extent of absorption or bioavailability. CONCLUSION: FP-025 was well tolerated and showed a favorable pharmacokinetic profile following ASD-in-Capsule dosing. Efficacy studies in target patient populations, including asthma, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis, are warranted. TRIAL REGISTRATION NUMBER: www.clinicaltrials.gov : NCT02238834 (Study I); NCT03304964 (Study II). Trial registration date: Study I was registered on 12 September 2014 while study II was registered on 9 October 2017.


Matrix Metalloproteinase 12/drug effects , Matrix Metalloproteinase Inhibitors/administration & dosage , Adult , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Matrix Metalloproteinase Inhibitors/adverse effects , Matrix Metalloproteinase Inhibitors/pharmacokinetics , Young Adult
2.
Int J Cardiol ; 185: 198-208, 2015 Apr 15.
Article En | MEDLINE | ID: mdl-25797678

RATIONALE: Matrix metalloproteinases (MMPs) regulate remodeling of the left ventricle (LV) post-myocardial infarction (MI). MMP-12 has potent macrophage-dependent remodeling properties in the atherosclerotic plaque; however, post-MI roles have not been examined. OBJECTIVE: The goal was to determine MMP-12 post-MI mechanisms. METHODS AND RESULTS: Male C57BL/6J mice (3-6 months old) were subjected to left coronary artery ligation. Saline or the RXP 470.1 MMP-12 inhibitor (MMP-12i; 0.5mg/kg/day) was delivered by osmotic mini-pump beginning 3h post-MI, and mice were sacrificed at day (d)1, 3, 5 or 7 post-MI and compared to d0 controls (mice without MI; n=6-12/group/time). MMP-12 expression increased early post-MI, and contrary to expected, neutrophils were a surprising early cellular source for MMP-12. MMP-12i reduced MMP-12 activity 33 ± 1% at d1 post-MI. Despite similar infarct areas and survival rates, MMP-12i led to greater LV dilation and worsened LV function. At d7 post-MI, MMP-12i prolonged pro-inflammatory cytokine upregulation (IL1r1, IL6ra, IL11, and Cxcr5) and decreased CD44 (both gene and protein levels). Hyaluronan (HA), a CD44 ligand, was elevated at d1 and d7 post-MI with MMP12i, as a result of decreased fragmentation. Because CD44-HA regulates neutrophil removal, apoptosis markers were evaluated. Caspase 3 increased, while cleaved caspase 3 levels decreased in MMP-12i group at d7 post-MI, indicating reduced neutrophil apoptosis. In isolated neutrophils, active MMP-12 directly stimulated CD44, caspase 3, and caspase 8 expression. CONCLUSION: Our results reveal a novel protective mechanism for MMP-12 in neutrophil biology. Post-MI, MMP-12i impaired CD44-HA interactions to suppress neutrophil apoptosis and prolong inflammation, which worsened LV function.


Gene Expression Regulation , Inflammation/genetics , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Myocardial Infarction/complications , Ventricular Dysfunction, Left/genetics , Ventricular Function, Left/physiology , Animals , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Inflammation/enzymology , Male , Matrix Metalloproteinase 12/biosynthesis , Matrix Metalloproteinase 12/drug effects , Mice , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/enzymology , RNA/genetics , Real-Time Polymerase Chain Reaction , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/drug effects , Ventricular Remodeling/physiology
3.
J Med Chem ; 56(3): 1149-59, 2013 Feb 14.
Article En | MEDLINE | ID: mdl-23343195

The molecular determinants responsible for the potency of the RXP470.1 phosphinic peptide inhibitor toward matrix metalloprotease-12 (MMP-12) remain elusive. To address this issue, structure-activity study, X-ray crystallography, and isothermal titration calorimetry (ITC) experiments were performed. The crystal structure of MMP-12/inhibitor complex (1.15 Å) reveals that the inhibitor establishes multiple interactions with the MMP-12 active site, with its long P(1)' side chain filling most of the S(1)' deep cavity. ITC experiments indicate that the binding of this inhibitor to MMP-12 is mostly entropy driven (ΔG° = -13.1 kcal/mol, ΔH° = -2.53 kcal/mol, and -TΔS° = -10.60 kcal/mol) and involves a proton uptake from the buffer. Comparing phosphinic versus hydroxamate inhibitors reveals that the chelation of the zinc ion is slightly different, leading the inhibitor backbone to adopt a position in which the hydrogen bonding with the MMP-12 active site is less favorable in phosphinic inhibitor while maintaining high affinity.


Matrix Metalloproteinase 12/drug effects , Protease Inhibitors/pharmacology , Crystallography, X-Ray , Models, Molecular , Protease Inhibitors/chemistry , Thermodynamics
4.
Asian Pac J Cancer Prev ; 13(4): 1657-62, 2012.
Article En | MEDLINE | ID: mdl-22799384

Fucosyltransferase IV (FUT4) has been implicated in cell adhesion, motility, and tumor progression in human epidermoid carcinoma A431 cells. We previously reported that it promotes cell proliferation through the ERK/MAPK and PI3K/Akt signaling pathways; however, the molecular mechanisms underlying FUT4- induced cell invasion remain unknown. In this study we determined the effect of FUT4 on expression of matrix metalloproteinase (MMP)-12 induced by EGF in A431 cells. Treatment with EGF resulted in an alteration of cell morphology and induced an increase in the expression of MMP-12. EGF induced nuclear translocation of nuclear factor κB (NF-κB) and resulted in phosphorylation of IκBα in a time-dependent manner. In addition, ERK1/2 and p38 MAPK were shown to play a crucial role in mediating EGF-induced NF-κB translocation and phosphorylation of IκBα when treated with the MAPK inhibitors, PD98059 and SB203580, which resulted in increased MMP-12 expression. Importantly, we showed that FUT4 up-regulated EGF-induced MMP-12 expression by promoting the phosphorylation of ERK1/2 and p38 MAPK, thereby inducing phosphorylation/ degradation of IκBα, NF-κB activation. Base on our data, we propose that FUT4 up-regulates expression of MMP-12 via a MAPK-NF-κB-dependent mechanism.


Carcinoma, Squamous Cell/enzymology , Epidermal Growth Factor/pharmacology , Fucosyltransferases/metabolism , Lewis X Antigen/metabolism , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 12/metabolism , NF-kappa B/metabolism , Analysis of Variance , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Nucleus/metabolism , Cytosol/metabolism , Fucosyltransferases/genetics , Gene Expression Regulation, Neoplastic , Humans , I-kappa B Proteins/metabolism , Lewis X Antigen/genetics , Matrix Metalloproteinase 12/drug effects , Matrix Metalloproteinase 12/genetics , NF-kappa B/drug effects , NF-kappa B p50 Subunit/metabolism , Protein Transport/drug effects , Transcription Factor RelA/metabolism , Transfection
5.
Toxicol Pathol ; 38(3): 402-15, 2010 Apr.
Article En | MEDLINE | ID: mdl-20215583

A short-term 5-day cigarette smoke exposure study was conducted in Fischer 344 rats to identify smoke-induced lung protein changes. Groups of 10 male and 10 female rats at 5 weeks of age were randomly assigned to one of four exposure groups. Animals received filtered air (control) or 75, 200, or 400 mg total particulate matter (TPM)/m(3) of diluted Kentucky reference 3R4F cigarette smoke. Nose-only exposures were conducted for 3 hours/day for 5 consecutive days. Mean body weights were significantly reduced only in male rats exposed to 400 mg TPM/m(3). Body weight gains were significantly reduced in 200- and 400-mg TPM/m(3)-exposed males and in all smoke-exposed females compared with controls. Alveolar histiocytosis increased slightly in all smoke exposed-females and 200- and 400-mg TPM/m(3)-exposed males. Cyclooxygenase-2 staining increased at 400 mg TPM/m(3). Matrix metalloproteinase-12 staining of alveolar macrophages and bronchiolar epithelia increased in smoke-exposed animals, especially 400-mg TPM/m(3)-exposed females. Protein kinase C-alpha staining increased in macrophages at 200- and 400-mg TPM/m(3) doses. c-Jun NH(2)-terminal kinases staining decreased in smoke-exposed tissues. The identified changed proteins play roles in inflammation, transformation, proliferation, stress activation, and apoptosis.


Lung/drug effects , Proteins/drug effects , Smoking/adverse effects , Tobacco Smoke Pollution/adverse effects , Animals , Body Weight/drug effects , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/drug effects , Female , Immunohistochemistry , Lung/metabolism , Lung/pathology , Male , Matrix Metalloproteinase 12/biosynthesis , Matrix Metalloproteinase 12/drug effects , Protein Kinase C/biosynthesis , Protein Kinase C/drug effects , Rats , Rats, Inbred F344 , Smoking/pathology , Time
6.
Immunology ; 128(1 Suppl): e315-24, 2009 Sep.
Article En | MEDLINE | ID: mdl-19019090

Formation of osteoclasts and consequent joint destruction are hallmarks of rheumatoid arthritis (RA). Here we show that LIGHT, a member of the tumour necrosis factor (TNF) superfamily, induced the differentiation into tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells (MNCs) of CD14(+) monocytes cocultured with nurse-like cells isolated from RA synovium, but not of freshly isolated CD14(+) monocytes. Receptor activator of nuclear factor-kappaB ligand (RANKL) enhanced this LIGHT-induced generation of TRAP-positive MNCs. The MNCs showed the phenotypical and functional characteristics of osteoclasts; they showed the expression of osteoclast markers such as cathepsin K, actin-ring formation, and the ability to resorb bone. Moreover, the MNCs expressed both matrix metalloproteinase 9 (MMP-9) and MMP-12, but the latter was not expressed in osteoclasts induced from CD14(+) monocytes by RANKL. Immunohistochemical analysis showed that the MMP-12-producing MNCs were present in the erosive areas of joints in RA, but not in the affected joints of osteoarthritic patients. These findings suggested that LIGHT might be involved in the progression of inflammatory bone destruction in RA, and that osteoclast progenitors might become competent for LIGHT-mediated osteoclastogenesis via interactions with synoviocyte-like nurse-like cells.


Arthritis, Rheumatoid/immunology , Monocytes/immunology , Osteoclasts/immunology , Synovial Membrane/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Acid Phosphatase/drug effects , Acid Phosphatase/immunology , Acid Phosphatase/metabolism , Arthritis, Rheumatoid/metabolism , Bone Resorption/immunology , Bone Resorption/metabolism , Bone and Bones/drug effects , Bone and Bones/immunology , Bone and Bones/metabolism , Bone and Bones/pathology , Cathepsin K/drug effects , Cathepsin K/immunology , Cathepsin K/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Humans , Isoenzymes/drug effects , Isoenzymes/immunology , Isoenzymes/metabolism , Matrix Metalloproteinase 12/drug effects , Matrix Metalloproteinase 12/immunology , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , RANK Ligand/pharmacology , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Tartrate-Resistant Acid Phosphatase , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
7.
Am J Cardiol ; 100(1): 23-7, 2007 Jul 01.
Article En | MEDLINE | ID: mdl-17599435

Several matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) have been implicated in the development and outcome of coronary artery disease (CAD). We investigated whether MMP-12 and TIMP-1 levels were associated with risk, severity, and outcome of CAD. Plasma MMP-12 and TIMP-1 levels are measured in 50 and 44 patients with CAD, respectively, by enzyme-linked immunosorbent assay. Of all patients, 16 were taking statins. Patients who were not on statins were classified into 3 groups according to number of >50% stenotic vessels. Compared with 29 volunteers without CAD, patients without statins (n = 34) had higher MMP-12 concentrations (1.71 vs 1.08 ng/ml, p = 0.021). MMP-12 levels were significantly lower in patients with than in those without statin treatment (0.99 vs 1.71 ng/ml, p = 0.008). There was no association between MMP-12 levels and number of >50% stenotic vessels. MMP-12 concentrations were not associated with outcome of CAD. However, plasma TIMP-1 levels were associated with restenosis independently of number of stenotic vessels and age (p = 0.035) but not with risk or severity of CAD. In conclusion, plasma MMP-12 concentration was associated with the presence of CAD. Statin therapy decreases plasma MMP-12 levels in patients with CAD. Increased TIMP-1 levels may prevent restenosis after angioplasty.


Coronary Artery Disease/blood , Matrix Metalloproteinase 12/blood , Tissue Inhibitor of Metalloproteinase-1/blood , Adult , Aged , Biomarkers , Case-Control Studies , Coronary Artery Disease/drug therapy , Coronary Restenosis/prevention & control , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Matrix Metalloproteinase 12/drug effects , Middle Aged , Prospective Studies , Risk Factors , Severity of Illness Index
8.
Expert Rev Cardiovasc Ther ; 5(2): 265-82, 2007 Mar.
Article En | MEDLINE | ID: mdl-17338671

Atherosclerotic plaque rupture, with subsequent occlusive thrombosis, is the underlying cause of most cases of sudden cardiac death. Matrix metalloproteinases (MMPs) are thought to mediate the progression of stable atherosclerotic lesions to an unstable phenotype that is prone to rupture through the destruction of strength-giving extracellular matrix (ECM) proteins. Smooth muscle cells secrete and deposit ECM proteins and are, therefore, considered protective against atherosclerotic plaque destabilization. However, similar to inflammatory cells (e.g., macrophages), smooth muscle cells release numerous MMPs that are capable of digesting ECM proteins. Thus, the interaction of smooth muscle cells and MMPs in atherosclerotic plaques is complex and not fully understood. Recently, research into the roles of MMPs and their endogenous inhibitors (tissue inhibitors of metalloproteinases), and their effects on smooth muscle behavior during plaque destabilization has been aided by the development of reproducible animal models of plaque instability. A plethora of studies has demonstrated that MMPs directly modulate smooth muscle behavior with both beneficial and deleterious effects on atherosclerotic plaque stability, in addition to their canonical effects on ECM remodeling. Consequently, broad-spectrum MMP inhibition may inhibit plaque-stabilizing mechanisms, such as smooth muscle cell growth, while conversely retarding ECM destruction and subsequent rupture. Hence the development of selective MMP inhibitors, that spare inhibitory effects on smooth muscle cell function, may be useful therapies to prevent plaque rupture and in this regard MMP-12 appears to be a particularly attractive target.


Atherosclerosis/pathology , Matrix Metalloproteinases/metabolism , Muscle, Smooth, Vascular/metabolism , Tissue Inhibitor of Metalloproteinases/pharmacology , Animals , Atherosclerosis/physiopathology , Cells, Cultured , Female , Humans , Male , Matrix Metalloproteinase 12/drug effects , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinases/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth, Vascular/drug effects , Rabbits , Risk Factors , Sensitivity and Specificity
...