Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905635

ABSTRACT

The interaction between menin and histone-lysine N-methyltransferase 2A (KMT2A) is a critical dependency for KMT2A- or nucleophosmin 1 (NPM1)-altered leukemias and an emerging opportunity for therapeutic development. JNJ-75276617 is a novel, orally bioavailable, potent, and selective protein-protein interaction inhibitor of the binding between menin and KMT2A. In KMT2A-rearranged (KMT2A-r) and NPM1-mutant (NPM1c) AML cells, JNJ-75276617 inhibited the association of the menin-KMT2A complex with chromatin at target gene promoters, resulting in reduced expression of several menin-KMT2A target genes, including MEIS1 and FLT3. JNJ-75276617 displayed potent anti-proliferative activity across several AML and ALL cell lines and patient samples harboring KMT2A- or NPM1-alterations in vitro. In xenograft models of AML and ALL, JNJ-75276617 reduced leukemic burden and provided a significant dose-dependent survival benefit accompanied by expression changes of menin-KMT2A target genes. JNJ-75276617 demonstrated synergistic effects with gilteritinib in vitro in AML cells harboring KMT2A-r. JNJ-75276617 further exhibited synergistic effects with venetoclax and azacitidine in AML cells bearing KMT2A-r in vitro, and significantly increased survival in mice. Interestingly, JNJ-75276617 showed potent anti-proliferative activity in cell lines engineered with recently discovered mutations (MEN1M327I or MEN1T349M) that developed in patients refractory to the menin-KMT2A inhibitor revumenib. A co-crystal structure of menin in complex with JNJ-75276617 indicates a unique binding mode distinct from other menin-KMT2A inhibitors, including revumenib. JNJ-75276617 is being clinically investigated for acute leukemias harboring KMT2A or NPM1 alterations, as a monotherapy for relapsed/refractory (R/R) acute leukemia (NCT04811560), or in combination with AML-directed therapies (NCT05453903).

2.
Clin Transl Sci ; 17(3): e13742, 2024 03.
Article in English | MEDLINE | ID: mdl-38494922

ABSTRACT

Relapsed/refractory (r/r) acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) outcomes remain poor. A targeted cluster of differentiation (CD)33 × CD3 bispecific antibody, JNJ-67571244, was assessed to identify the maximum tolerated dose (MTD), recommended phase II dose (RP2D), safety and tolerability, and preliminary clinical activity in patients with r/rAML or r/rMDS. This first-in-human, open-label, phase I, dose-escalation/dose-expansion study included patients with r/rAML or r/rMDS who were ineligible for or had exhausted standard therapeutic options. JNJ-67571244 was administered intravenously or subcutaneously using step-up dosing until ≥1 discontinuation condition was met. Outcomes included safety/tolerability, preliminary clinical activity, and systemic pharmacokinetics and pharmacodynamics. The study was terminated after evaluating 10 dose-escalation cohorts (n = 68) and before starting dose-expansion. Overall, 11 (16.2%) patients experienced ≥1 dose-limiting toxicity; all experienced ≥1 treatment-emergent adverse event (TEAE; treatment related: 60 [88.2%]); and 64 (94.1%) experienced ≥1 TEAE of Grade ≥3 toxicity (treatment related: 28 [41.2%]). Although some patients had temporary disease burden reductions, no responses were seen. JNJ-67571244 administration increased multiple cytokines, which coincided with incidence of cytokine release syndrome, infusion-related reactions, and elevated liver function tests. A prolonged step-up strategy was tested to improve tolerability, though this approach did not prevent hepatotoxicity. T-cell activation following treatment suggested target engagement but did not correlate with clinical activity. Safely reaching the projected exposure level for JNJ-67571244 efficacy was not achieved, thus MTD and RP2D were not determined.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Sialic Acid Binding Ig-like Lectin 3/immunology
3.
Clin Transl Sci ; 16(3): 429-435, 2023 03.
Article in English | MEDLINE | ID: mdl-36564917

ABSTRACT

This study aimed to identify a recommended phase II dose and evaluate the safety, tolerability, pharmacokinetics/pharmacodynamics, and preliminary clinical activity of JNJ-63709178, a CD123/CD3 dual-targeting antibody, in patients with relapsed or refractory acute myeloid leukemia. Intravenous (i.v.) and subcutaneous (s.c.) administration of JNJ-63709178 were evaluated. The i.v. infusions were administered once every 2 weeks (cohorts 1-5 [n = 17]) or twice weekly (cohorts 6-11 [n = 36]). A twice-weekly s.c. dosing regimen with step-up dosing was also studied (s.c. cohorts 1-2 [n = 9]). Treatment-emergent adverse events (TEAEs) greater than or equal to grade 3 were observed in 11 (65%) patients in cohorts 1-5 and 33 (92%) patients in cohorts 6-11. At the highest i.v. dose (4.8 µg/kg), 5 (71%) patients discontinued treatment due to TEAEs. For s.c. administration (n = 9), eight (89%) patients experienced TEAEs greater than or equal to grade 3 and injection site reactions (≤ grade 3) emerged in all patients. At 4.8 µg/kg (i.v. and s.c.), the mean maximum serum concentrations were 30.3 and 3.59 ng/ml, respectively. Increases in multiple cytokines were observed following i.v. and s.c. administrations, and step-up dosing strategies did not mitigate cytokine production or improve the safety profile and led to limited duration of treatment. Minimal clinical activity was observed across all cohorts. The i.v. and s.c. dosing of JNJ-63709178 was associated with suboptimal drug exposure, unfavorable safety profiles, limited clinical activity, and inability to identify a recommended phase II dose.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , Interleukin-3 Receptor alpha Subunit/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy
4.
Clin Pharmacokinet ; 56(6): 607-615, 2017 06.
Article in English | MEDLINE | ID: mdl-27722854

ABSTRACT

INTRODUCTION: Elevated interleukin (IL)-6 occurs in patients with active rheumatoid arthritis (RA), which has been shown to lead to a decrease in cytochrome P450 (CYP) enzyme activity and alterations in drug concentrations metabolized by CYP. IL-6 signaling blockade by IL-6 receptor (IL-6R) antagonists may reverse this effect of IL-6 and restore CYP activity. This study evaluated the pharmacokinetic profile of simvastatin (a CYP3A4 substrate) before and 1 week after a single dose of sarilumab (a human monoclonal antibody [mAb] blocking the IL-6Rα) in patients with RA, to assess potential interaction. METHODS: Nineteen patients with active RA received oral simvastatin 40 mg 1 day before and 7 days after subcutaneous injection of sarilumab 200 mg. The pharmacokinetic parameters of simvastatin and its primary metabolite, ß-hydroxy-simvastatin acid, were calculated using noncompartmental analysis. RESULTS: Compared with simvastatin alone, single-dose simvastatin administration 7 days after single-dose sarilumab administration in patients with RA resulted in reduced simvastatin and ß-hydroxy-simvastatin acid exposure in plasma. Mean effect ratios (90 % confidence interval) for simvastatin peak plasma concentration (C max) and area under the concentration-time curve extrapolated to infinity (AUC∞) were 54.1 % (42.2-69.4 %) and 54.7 % (47.2-63.3 %), respectively. No changes occurred in time to C max or half-life for either simvastatin or ß-hydroxy-simvastatin acid after sarilumab administration. CONCLUSIONS: Sarilumab treatment resulted in a reduction in exposure of simvastatin, consistent with reversal of IL-6-mediated CYP3A4 suppression in patients with active RA, as was reported for tocilizumab with simvastatin and for sirukumab with midazolam. CLINICAL TRIAL REGISTRATION NUMBER: NCT02017639.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/metabolism , Cytochrome P-450 CYP3A/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Interleukin-6/antagonists & inhibitors , Simvastatin/pharmacokinetics , Adult , Aged , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Drug Interactions , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Interleukin-6/blood , Male , Middle Aged , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/blood , Simvastatin/analogs & derivatives , Simvastatin/blood
5.
J Clin Pharmacol ; 52(10): 1506-15, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22128201

ABSTRACT

Clopidogrel requires CYP450-mediated hepatic metabolism to form its active metabolite (clopi-H4). This randomized, placebo-controlled, crossover study was designed to characterize the effect of a high-fat or standard breakfast on adenosine diphosphate (ADP)-induced platelet aggregation and exposure to unchanged clopidogrel and clopi-H4 following clopidogrel (300-mg loading dose, 75 mg/d for 4 days) in 72 healthy men. At day 5 and as assessed by liquid chromatography-tandem mass spectrometry, unchanged clopidogrel area under the concentration- time curve from 0 to 24 hours (AUC(0-24)) increased 3.32-fold (90% confidence interval [CI], 2.88-3.84), and clopi-H4 AUC(0-24) decreased nonsignificantly by 12% (90% CI, 0.82-0.94) upon administration of clopidogrel with a standard breakfast. The estimated treatment difference in maximum platelet aggregation (MPA) induced by ADP 5 µM and assessed by light transmission aggregometry was 4.7%, with the 90% CI (0.9%-8.5%) contained within the prespecified equipotency range of ±15%. The mean ± standard deviation of day 5 inhibition of platelet aggregation was 49.7% ± 17.2% and 54.0% ± 13.3% in the fed and fasted states, respectively. Despite increased unchanged clopidogrel and slightly decreased clopi-H4 exposure following clopidogrel administration, the numerical increase in MPA in the fed versus fasted state was small and within the prespecified limit of equipotency. These findings confirm that clopidogrel can be taken with or without food.


Subject(s)
Food-Drug Interactions , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Ticlopidine/analogs & derivatives , Adenosine Diphosphate , Adult , Area Under Curve , Aryl Hydrocarbon Hydroxylases/genetics , Breakfast , Clopidogrel , Cross-Over Studies , Cytochrome P-450 CYP2C19 , Diet, High-Fat , Genotype , Humans , Male , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Ticlopidine/administration & dosage , Ticlopidine/blood , Ticlopidine/pharmacokinetics , Young Adult
6.
Semin Nephrol ; 26(2): 89-94, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16530601

ABSTRACT

Focal and segmental glomerulosclerosis (FSGS) is a pathologic entity that is a common and increasing cause of end-stage renal disease. Typical manifestations include proteinuria, hypertension, worsening renal insufficiency, and, frequently, renal failure. The etiology, however, remains unknown in a majority of patients. There is an estimated recurrence rate of 30% to 40% in renal transplant patients, suggesting that the pathogenesis is not solely a result of intrinsic kidney disease. Although some of its characteristics have been reported, the precise identification of a circulating factor associated with FSGS has not been made. Remarkable progress has been made in recent years regarding biologic mechanisms surrounding FSGS and proteinuria. Insight into the pathogenesis of FSGS has been gained through the study of hereditary forms of FSGS and nephrotic syndromes. Mutations in cytoskeletal proteins that affect podocyte structure have been the target until recently. Here we review the current understanding of this glomerular disease and areas for future concentration.


Subject(s)
Biomarkers/blood , Glomerulosclerosis, Focal Segmental , Adaptor Proteins, Signal Transducing , Cytoskeletal Proteins/blood , Disease Progression , Glomerulosclerosis, Focal Segmental/complications , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Intracellular Signaling Peptides and Proteins , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/pathology , Membrane Proteins/blood , Risk Factors , TRPC Cation Channels/blood , TRPC6 Cation Channel , src Homology Domains
8.
Science ; 308(5729): 1801-4, 2005 Jun 17.
Article in English | MEDLINE | ID: mdl-15879175

ABSTRACT

Focal and segmental glomerulosclerosis (FSGS) is a kidney disorder of unknown etiology, and up to 20% of patients on dialysis have been diagnosed with it. Here we show that a large family with hereditary FSGS carries a missense mutation in the TRPC6 gene on chromosome 11q, encoding the ion-channel protein transient receptor potential cation channel 6 (TRPC6). The proline-to-glutamine substitution at position 112, which occurs in a highly conserved region of the protein, enhances TRPC6-mediated calcium signals in response to agonists such as angiotensin II and appears to alter the intracellular distribution of TRPC6 protein. Previous work has emphasized the importance of cytoskeletal and structural proteins in proteinuric kidney diseases. Our findings suggest an alternative mechanism for the pathogenesis of glomerular disease.


Subject(s)
Calcium Channels/genetics , Glomerulosclerosis, Focal Segmental/genetics , Mutation, Missense , Amino Acid Substitution , Angiotensin II/metabolism , Angiotensin II/pharmacology , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium Signaling , Carbachol/pharmacology , Cell Line , Cell Membrane/metabolism , Chromosomes, Human, Pair 11/genetics , Exons , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Haplotypes , Humans , Kidney/metabolism , Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Male , Patch-Clamp Techniques , Pedigree , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Sequence Analysis, DNA , Sodium/metabolism , TRPC Cation Channels , TRPC6 Cation Channel , Transfection , Uridine Triphosphate/metabolism , Uridine Triphosphate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...