Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACR Open Rheumatol ; 6(4): 205-213, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38311369

ABSTRACT

OBJECTIVE: To assess the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple injections of M6495, a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5)  nanobody, in healthy volunteers and patients with osteoarthritis. METHODS: Two randomized, placebo-controlled, double-blind studies were performed. Study 1 enrolled 54 healthy male volunteers who received one subcutaneous (s.c.) injection of M6495 (1-300 mg) or placebo (ratio 2:1), evaluating safety, PK, and PD as changes in the serum aggrecan fragment alanine-arginine-glycine-serine (ARGS). Study 2 enrolled 32 patients with osteoarthritis with Kellgren-Lawrence grades 2 to 4 and pain greater than or equal to 40 on the Western Ontario and McMaster Universities Arthritis Index pain subscale at screening and evaluated the safety, PK, and PD of three doses every two weeks (75-300 mg per dose) or six once-weekly M6495 s.c. doses (300 mg) or placebo (ratio 3:1) over 106 days' follow-up. RESULTS: M6495 in single and multiple doses of less than or equal to 300 mg s.c. weekly was well tolerated with no clinically significant changes in any safety parameter. Adverse events more frequently reported in the M6495 groups were mostly mild cases of injection site reactions, myalgia, and nausea, which resolved after treatment cessation. The elimination half-life of single s.c. doses of M6495 ranged from 79 to 267 hours. M6495 administration substantially reduced serum ARGS levels, indicative of target engagement and indicating disease-modifying potential of M6495. CONCLUSION: Treatment with M6495 in single and multiple doses up to and including 300 mg s.c. was found to be well tolerated and adequately safe for further clinical evaluation of potential disease-modifying effects.

2.
Pharmacol Res Perspect ; 9(5): e00842, 2021 10.
Article in English | MEDLINE | ID: mdl-34414672

ABSTRACT

This study evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of single and multiple oral doses of enpatoran (formerly named M5049), a new toll-like receptor (TLR) 7 and 8 dual antagonist, and the effect of food on a single dose in healthy participants. In this single phase 1, randomized (3:1), double-blind, placebo-controlled study, 96 participants received single and multiple ascending oral doses of enpatoran. Participants in single-dose cohorts received one dose of enpatoran (1, 3, 9, 25, 50, 100, or 200 mg) or placebo using a sentinel dosing strategy. Multiple-dose cohorts received enpatoran (9, 25, or 200 mg once daily, or 25 or 50 mg twice daily) or placebo for 14 days. Safety, tolerability, PK, and PD (ex vivo-stimulated cytokine secretion) were assessed in both parts. The effect of food was assessed in an open-label, one-way crossover study in the 25 mg single-dose cohort. Single- and multiple-oral doses of enpatoran up to 200 mg were well tolerated and no significant dose-limiting adverse events or safety signals were observed under fasting or fed conditions. PK parameters were linear and dose-proportional across the dose range evaluated, with a slightly delayed absorption and lower peak concentration observed at 25 mg with food. Exposure-dependent inhibition of ex vivo-stimulated interleukin-6 secretion was observed, with maximum inhibition at 200 mg. Enpatoran was well tolerated at doses up to 200 mg. Further investigation of enpatoran is warranted as a potential treatment for diseases driven by TLR7/8 overactivation, such as systemic lupus erythematosus and COVID-19 pneumonia.


Subject(s)
Immunologic Factors/pharmacology , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 8/antagonists & inhibitors , Administration, Oral , Adult , COVID-19/immunology , Double-Blind Method , Female , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Male , SARS-CoV-2 , COVID-19 Drug Treatment
3.
Ann Rheum Dis ; 80(8): 1062-1069, 2021 08.
Article in English | MEDLINE | ID: mdl-33962962

ABSTRACT

OBJECTIVE: The FORWARD (FGF-18 Osteoarthritis Randomized Trial with Administration of Repeated Doses) trial assessed efficacy and safety of the potential disease-modifying osteoarthritis drug (DMOAD) sprifermin in patients with knee osteoarthritis. Here, we report 5-year efficacy and safety results. METHODS: Patients were randomised to intra-articular sprifermin 100 µg or 30 µg every 6 months (q6mo) or 12 months, or placebo, for 18 months. The primary analysis was at year 2, with follow-up at years 3, 4 and 5. Additional post hoc exploratory analyses were conducted in patients with baseline minimum radiographic joint space width 1.5-3.5 mm and Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain 40-90, a subgroup at risk (SAR) of progression. RESULTS: 378 (69%) patients completed the 5-year follow-up. A significant dose-response in total femorotibial joint cartilage thickness with sprifermin (trend test, p<0.001) and a 0.05 mm mean difference with sprifermin 100 µg q6mo versus placebo (95% CI 0.00 to 0.10; p=0.015) were sustained to year 5. WOMAC pain scores improved ~50% from baseline in all groups. No patient in the 100 µg q6mo group had replacement of the treated knee. 96%-98% of patients receiving sprifermin and 98% placebo reported adverse events, most were mild or moderate and deemed unrelated to treatment. Adverse event-related study withdrawals were <10%. Differentiation in WOMAC pain between sprifermin 100 µg q6mo and placebo in the SAR (n=161) at year 3 was maintained to year 5 (-10.08; 95% CI -25.68 to 5.53). CONCLUSION: In the longest DMOAD trial reported to date, sprifermin maintained long-term structural modification of articular cartilage over 3.5 years post-treatment. Potential translation to clinical benefit was observed in the SAR. TRIAL REGISTRATION NUMBER: NCT01919164.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Double-Blind Method , Fibroblast Growth Factors , Humans , Injections, Intra-Articular , Magnetic Resonance Imaging , Osteoarthritis, Knee/drug therapy , Pain/drug therapy , Treatment Outcome
4.
Rheumatology (Oxford) ; 60(11): 5379-5389, 2021 11 03.
Article in English | MEDLINE | ID: mdl-33547784

ABSTRACT

OBJECTIVES: Atacicept reduced SLE disease activity in the phase 2b ADDRESS II study, particularly in patients with high disease activity (HDA; SLEDAI-2K ≥10) at screening. We assessed long-term safety and efficacy of atacicept in the long-term extension (LTE) of ADDRESS II. METHODS: In the 24-week, randomized, double-blind, placebo-controlled ADDRESS II study, patients received weekly atacicept (75 or 150 mg) or placebo. Atacicept was continued at the same dose in atacicept-treated patients in the LTE; placebo-treated patients switched to atacicept 150 mg. Long-term safety was the primary endpoint. Secondary endpoints included SLE responder index (SRI)-4 and SRI-6 response rates and flares. RESULTS: In total, 253 patients entered the ADDRESS II LTE; 88 received atacicept 150 mg, 82 atacicept 75 mg and 83 placebo/atacicept 150 mg. Median active treatment duration in the LTE was 83.8 weeks. Frequencies of treatment-emergent adverse events (TEAEs) were similar across groups (90.4-93.2%), and 12.5%, 14.6% and 21.7% of patients in the atacicept 150 mg, atacicept 75 mg and placebo/atacicept 150 mg groups reported serious TEAEs during the treatment period. The proportions of patients with TEAEs leading to discontinuation were 5.7%, 4.9% and 10.8%, respectively. SRI-4 and SRI-6 response rates were maintained with atacicept in the modified intent-to-treat and HDA populations and those on continuous 150 mg had a reduced risk of first severe flare and longer time to first severe flare vs those who initially received placebo. CONCLUSION: Long-term treatment with atacicept 150 mg in SLE patients had an acceptable safety profile, with durable efficacy. TRIAL REGISTRATION: ClinicalTrials.gov, http://clinicaltrials.gov, NCT02070978.


Subject(s)
Lupus Erythematosus, Systemic/drug therapy , Recombinant Fusion Proteins/therapeutic use , Adult , Biomarkers/blood , Double-Blind Method , Female , Humans , Lupus Erythematosus, Systemic/blood , Male , Middle Aged , Remission Induction , Severity of Illness Index , Symptom Flare Up , Treatment Outcome
5.
BMC Surg ; 14: 107, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25511186

ABSTRACT

BACKGROUND: Primary mesenchymal chondrosarcoma of the kidney is an extremely rare malignant tumor. To our best knowledge, only 9 such cases have been reported so far. CASE PRESENTATION: In the current paper, we present the case of a 67 year-old patient with recurrent left lumbar pain, increased fatigability and intermittent macroscopic hematuria. He underwent a surgical resection of the left kidney and left hemicolon.The pathological diagnosis was primary extraskeletal renal mesenchymal chondrosarcoma. Overall survival was 9 months, with pulmonary metastasis and local recurrence at 6 months. The management of the patient is described, from the initial differential diagnosis, after the first clinical examination to the surgical resection, with a special emphasis on the surgical procedures that were carried out. CONCLUSION: Extraskeletal chondrosarcoma of primary origin in the kidney are extremely rare tumors with a highly malignant potential and very poor prognosis. Because the role of chemotherapy or radiation therapy has not been evaluated properly yet, we underline the importance of surgery in the management of such cases as the main and best approach to achieve clinical remission and long-term survival, provided the patient is referred to a surgical consult in time.


Subject(s)
Chondrosarcoma, Mesenchymal/surgery , Kidney Neoplasms/surgery , Aged , Chondrosarcoma, Mesenchymal/secondary , Diagnosis, Differential , Fatal Outcome , Humans , Kidney Neoplasms/pathology , Lung Neoplasms/secondary , Male , Neoplasm Recurrence, Local
7.
Am J Respir Crit Care Med ; 177(1): 56-65, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17916809

ABSTRACT

RATIONALE: Transforming growth factor (TGF)-beta has a central role in driving many of the pathological processes that characterize pulmonary fibrosis. Inhibition of the integrin alpha(v)beta6, a key activator of TGF-beta in lung, is an attractive therapeutic strategy, as it may be possible to inhibit TGF-beta at sites of alpha(v)beta6 up-regulation without affecting other homeostatic roles of TGF-beta. OBJECTIVES: To analyze the expression of alpha(v)beta6 in human pulmonary fibrosis, and to functionally test the efficacy of therapeutic inhibition of alpha(v)beta6-mediated TGF-beta activation in murine bleomycin-induced pulmonary fibrosis. METHODS: Lung biopsies from patients with a diagnosis of systemic sclerosis or idiopathic pulmonary fibrosis were stained for alpha(v)beta6 expression. A range of concentrations of a monoclonal antibody that blocks alpha(v)beta6-mediated TGF-beta activation was evaluated in murine bleomycin-induced lung fibrosis. MEASUREMENTS AND MAIN RESULTS: Alpha(v)beta6 is overexpressed in human lung fibrosis within pneumocytes lining the alveolar ducts and alveoli. In the bleomycin model, alpha(v)beta6 antibody was effective in blocking pulmonary fibrosis. At high doses, there was increased expression of markers of inflammation and macrophage activation, consistent with the effects of TGF-beta inhibition in the lung. Low doses of antibody attenuated collagen expression without increasing alveolar inflammatory cell populations or macrophage activation markers. CONCLUSIONS: Partial inhibition of TGF-beta using alpha(v)beta6 integrin antibodies is effective in blocking murine pulmonary fibrosis without exacerbating inflammation. In addition, the elevated expression of alpha(v)beta6, an activator of the fibrogenic cytokine, TGF-beta, in human pulmonary fibrosis suggests that alpha(v)beta6 monoclonal antibodies could represent a promising new therapeutic strategy for treating pulmonary fibrosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Integrins/antagonists & inhibitors , Pulmonary Fibrosis/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antigens, Neoplasm/physiology , Collagen/metabolism , Dose-Response Relationship, Drug , Integrins/physiology , Mice , Mice, Inbred BALB C , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/immunology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/therapy , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Scleroderma, Systemic/therapy , Transforming Growth Factor beta/physiology
8.
J Neurochem ; 87(5): 1184-92, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14622098

ABSTRACT

Caspases play an important role in neurodegeneration in Huntington's disease (HD). Members of the Bcl-2 family are critical modulators of terminal cell death pathways. However, alterations of Bcl-2 family members and their functional role in an in vivo model of HD have not been documented. With the goal of gaining mechanistic insight, we used a transgenic mouse model of HD (R6/2) to investigate the chronology of caspase activation and functional alterations in members of the Bcl-2 family. In R6/2 mice caspase activation precedes proapoptotic changes in Bcl-2 family members. Of the caspases that we screened, caspase-1-like activation was the first to be detected in the disease process (7 weeks). Proapoptotic changes in members of the Bcl-2 family were first detected at 9 weeks. To demonstrate a potential functional/therapeutic role of Bcl-2 in HD, we crossed R6/2 mice with mice overexpressing Bcl-2 in neurons. Transgenic expression of Bcl-2 in R6/2 mice resulted in slight prolonged survival. Understanding the chronology of apoptotic events provides important information for appropriate therapeutic targeting in this devastating and untreatable disease.


Subject(s)
Apoptosis , Caspases/metabolism , Huntington Disease/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/physiology , Age Factors , Animals , Blotting, Western , Crosses, Genetic , Disease Models, Animal , Disease Progression , Fluorescent Antibody Technique , Gene Expression , Huntington Disease/genetics , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics
9.
Nature ; 417(6884): 74-8, 2002 May 02.
Article in English | MEDLINE | ID: mdl-11986668

ABSTRACT

Minocycline mediates neuroprotection in experimental models of neurodegeneration. It inhibits the activity of caspase-1, caspase-3, inducible form of nitric oxide synthetase (iNOS) and p38 mitogen-activated protein kinase (MAPK). Although minocycline does not directly inhibit these enzymes, the effects may result from interference with upstream mechanisms resulting in their secondary activation. Because the above-mentioned factors are important in amyotrophic lateral sclerosis (ALS), we tested minocycline in mice with ALS. Here we report that minocycline delays disease onset and extends survival in ALS mice. Given the broad efficacy of minocycline, understanding its mechanisms of action is of great importance. We find that minocycline inhibits mitochondrial permeability-transition-mediated cytochrome c release. Minocycline-mediated inhibition of cytochrome c release is demonstrated in vivo, in cells, and in isolated mitochondria. Understanding the mechanism of action of minocycline will assist in the development and testing of more powerful and effective analogues. Because of the safety record of minocycline, and its ability to penetrate the blood-brain barrier, this drug may be a novel therapy for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Cytochrome c Group/metabolism , Minocycline/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Age of Onset , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Caspases/metabolism , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Progression , Enzyme Activation/drug effects , Humans , Infarction, Middle Cerebral Artery , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mitochondria/enzymology , Mitochondrial Swelling/drug effects , N-Methylaspartate/toxicity , Permeability/drug effects , Rats , Survival Rate , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...