Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Publication year range
1.
Blood ; 2024 05 20.
Article in English | MEDLINE | ID: mdl-38768337

ABSTRACT

Estimating progression-free survival (PFS) and overall survival (OS) superiority during clinical trials of multiple myeloma (MM) has become increasingly challenging as novel therapeutics have improved patient outcomes. Thus, it is imperative to identify earlier endpoint surrogates that are predictive of long-term clinical benefit to expedite development of more effective therapies. Minimal residual disease (MRD)-negativity is a common intermediate endpoint that has shown prognostic value for clinical benefit in trials of patients with multiple myeloma (MM). This meta-analysis was based on the FDA guidance for considerations for a meta-analysis of MRD as a clinical endpoint and evaluates MRD-negativity as an early endpoint reasonably likely to predict long-term clinical benefit. Eligible studies were phase 2 or 3 randomized controlled clinical trials measuring MRD negativity as an endpoint in patients with MM, with follow-up of ≥6 months following an a priori defined time point of 12±3 months post-randomization. Eight newly diagnosed MM-(NDMM)-studies evaluating 4,907 patients were included. Trial-level associations between MRD-negativity and PFS were R2WLSiv (95% CI) 0.67 (0.43-0.91) and R2copula 0.84 (0.64->0.99) at the 12-month timepoint. The individual-level association between 12-month MRD negativity and PFS resulted in a global odds ratio of 4.02 (95% CI: 2.57-5.46). For relapse/refractory MM-(RRMM), there were four studies included, and the individual-level association between 12-month MRD negativity and PFS resulted in a global odds ratio of 7.67 (4.24-11.10). A clinical trial demonstrating a treatment effect on MRD is reasonably likely to eventually demonstrate a treatment effect on PFS, suggesting that MRD may be an early clinical endpoint reasonably likely to predict clinical benefit in MM, that may be used to support accelerated approval and thereby expedite the availability of new drugs to patients with MM.

2.
Nature ; 488(7411): 394-8, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22810587

ABSTRACT

Cardiac hypertrophy is initiated as an adaptive response to sustained overload but progresses pathologically as heart failure ensues. Here we report that genetic loss of APJ, a G-protein-coupled receptor, confers resistance to chronic pressure overload by markedly reducing myocardial hypertrophy and heart failure. In contrast, mice lacking apelin (the endogenous APJ ligand) remain sensitive, suggesting an apelin-independent function of APJ. Freshly isolated APJ-null cardiomyocytes exhibit an attenuated response to stretch, indicating that APJ is a mechanosensor. Activation of APJ by stretch increases cardiomyocyte cell size and induces molecular markers of hypertrophy. Whereas apelin stimulates APJ to activate Gαi and elicits a protective response, stretch signals in an APJ-dependent, G-protein-independent fashion to induce hypertrophy. Stretch-mediated hypertrophy is prevented by knockdown of ß-arrestins or by pharmacological doses of apelin acting through Gαi. Taken together, our data indicate that APJ is a bifunctional receptor for both mechanical stretch and the endogenous peptide apelin. By sensing the balance between these stimuli, APJ occupies a pivotal point linking sustained overload to cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Receptors, G-Protein-Coupled/metabolism , Adipokines , Animals , Aorta/pathology , Apelin , Apelin Receptors , Arrestins/deficiency , Arrestins/genetics , Arrestins/metabolism , Blood Pressure , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mechanoreceptors/metabolism , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , beta-Arrestins
3.
Xenobiotica ; 48(9): 904-910, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29027832

ABSTRACT

1. Venetoclax is a novel, small molecule B-cell lymphoma-2 (BCL-2) inhibitor that has demonstrated clinical efficacy in a variety of haematological malignancies. Since venetoclax is an inhibitor of P glycoprotein (P-gp) transporter, a study was conducted in healthy, female volunteers to evaluate the effect of venetoclax on the pharmacokinetics of digoxin, a P-gp probe substrate. 2. Volunteers received a single oral dose of digoxin (0.5 mg) with or without a single oral dose of venetoclax (100  mg). Serial blood samples were obtained for pharmacokinetic assessments of digoxin and venetoclax and serial urine samples were obtained for measurement of digoxin concentrations. Safety was assessed throughout the study. 3. Coadministration of digoxin and venetoclax increased digoxin maximum observed plasma concentration (Cmax) by 35% and area under the plasma-concentration time curve (AUC0-∞) by 9%. Digoxin half-life, renal clearance and the fraction excreted unchanged in urine remained relatively similar. The results of this study indicate that venetoclax can increase the concentrations of P-gp substrates. Narrow therapeutic index P-gp substrates should be administered six hours prior to venetoclax to minimise the potential interaction.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Digoxin/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cross-Over Studies , Digoxin/administration & dosage , Drug Interactions , Female , Half-Life , Humans , Middle Aged , Sulfonamides/administration & dosage
4.
Hum Psychopharmacol ; 32(1)2017 01.
Article in English | MEDLINE | ID: mdl-27925283

ABSTRACT

Dopamine can modulate long-term episodic memory. Its potential role on the generation of false memories, however, is less well known. In a randomized, double-blind, placebo-controlled experiment, 24 young healthy volunteers ingested a 4-mg oral dose of haloperidol, a dopamine D2 -receptor antagonist, or placebo, before taking part in a recognition memory task. Haloperidol was active during both study and test phases of the experiment. Participants in the haloperidol group produced more false recognition responses than those in the placebo group, despite similar levels of correct recognition. These findings show that dopamine blockade in healthy volunteers can specifically increase false recognition memory.


Subject(s)
Dopamine Antagonists/pharmacology , Haloperidol/pharmacology , Memory/drug effects , Photic Stimulation/methods , Recognition, Psychology/drug effects , Repression, Psychology , Adult , Antipsychotic Agents/pharmacology , Double-Blind Method , Female , Healthy Volunteers/psychology , Humans , Male , Memory/physiology , Recognition, Psychology/physiology , Young Adult
5.
Circ Res ; 112(1): 48-56, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-22993413

ABSTRACT

RATIONALE: Mitogen-activated protein kinases (MAPKs) are activated in the heart by disease-inducing and stress-inducing stimuli, where they participate in hypertrophy, remodeling, contractility, and heart failure. A family of dual-specificity phosphatases (DUSPs) directly inactivates each of the MAPK terminal effectors, potentially serving a cardioprotective role. OBJECTIVE: To determine the role of DUSP1 and DUSP4 in regulating p38 MAPK function in the heart and the effect on disease. METHODS AND RESULTS: Here, we generated mice and mouse embryonic fibroblasts lacking both Dusp1 and Dusp4 genes. Although single nulls showed no molecular effects, combined disruption of Dusp1/4 promoted unrestrained p38 MAPK activity in both mouse embryonic fibroblasts and the heart, with no change in the phosphorylation of c-Jun N-terminal kinases or extracellular signal-regulated kinases at baseline or with stress stimulation. Single disruption of either Dusp1 or Dusp4 did not result in cardiac pathology, although Dusp1/4 double-null mice exhibited cardiomyopathy and increased mortality with aging. Pharmacological inhibition of p38 MAPK with SB731445 ameliorated cardiomyopathy in Dusp1/4 double-null mice, indicating that DUSP1/4 function primarily through p38 MAPK in affecting disease. At the cellular level, unrestrained p38 MAPK activity diminished cardiac contractility and Ca2+ handling, which was acutely reversed with a p38 inhibitory compound. Poor function in Dusp1/4 double-null mice also was partially rescued by phospholamban deletion. CONCLUSIONS: Our data demonstrate that Dusp1 and Dusp4 are cardioprotective genes that play a critical role in the heart by dampening p38 MAPK signaling that would otherwise reduce contractility and induce cardiomyopathy.


Subject(s)
Cardiomyopathies/enzymology , Dual Specificity Phosphatase 1/deficiency , Myocytes, Cardiac/enzymology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cardiomyopathies/prevention & control , Cells, Cultured , Disease Models, Animal , Dual Specificity Phosphatase 1/genetics , Enzyme Activation , Fibroblasts/enzymology , Gene Expression Regulation , Hemodynamics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatases , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Transl Oncol ; 37: 101767, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37657154

ABSTRACT

BACKGROUND: FDG PET/CT is a tool for assessing response to therapy in various cancers, and may provide an earlier biomarker of clinical response. We developed a novel semi-automated approach for analyzing FDG PET/CT images in patients with multiple myeloma (MM) to standardize FDG PET application. METHODS: Patients (n = 8) with relapsed/refractory MM from the Phase 2 study (NCT02899052) of venetoclax plus carfilzomib and dexamethasone underwent FDG PET/CT at baseline and up to two timepoints during treatment. Images were processed using an established automated segmentation algorithm, with the modification that a red marrow region in an unaffected lumbar vertebra was used to define background standardized uptake value normalized to lean body mass (SUL) threshold above which uptake was considered disease-specific uptake. This approach was compared to lesion segmentation, and to International Myeloma Working Group (IMWG) response criteria, including minimal residual disease (MRD). RESULTS: The two FDG PET analysis techniques agreed on evaluation of patient-level SULpeak for 67% of scans. In the metabolic response assessment per PET Response Criteria in Solid Tumors (PERCIST), the two techniques agreed in 75% of patients. Differences between techniques occurred in low-uptake lesions due to greater reader sensitivity to lesions with uptake marginally above background. PERCIST outcomes were generally in agreement with IMWC and MRD. CONCLUSIONS: This semi-automated analysis was in high agreement with standard approaches for detecting response to MM therapy. This proof-of-concept study suggests that larger studies should be conducted to confirm how FDG PET analysis may aid early response detection in MM.

7.
Cancer Res Commun ; 3(12): 2497-2509, 2023 12 08.
Article in English | MEDLINE | ID: mdl-37956312

ABSTRACT

The BCL2 inhibitor venetoclax promotes apoptosis in blood cancer cells and is approved for treatment of chronic lymphocytic leukemia and acute myeloid leukemia. However, multiple myeloma cells are frequently more dependent on MCL-1 for survival, conferring resistance to venetoclax. Here we report that mevalonate pathway inhibition with statins can overcome resistance to venetoclax in multiple myeloma cell lines and primary cells. In addition, statins sensitize to apoptosis induced by MCL-1 inhibitor, S63845. In retrospective analysis of venetoclax clinical studies in multiple myeloma, background statin use was associated with a significantly enhanced rate of stringent complete response and absence of progressive disease. Statins sensitize multiple myeloma cells to venetoclax by upregulating two proapoptotic proteins: PUMA via a p53-independent mechanism and NOXA via the integrated stress response. These findings provide rationale for prospective testing of statins with venetoclax regimens in multiple myeloma. SIGNIFICANCE: BH3 mimetics including venetoclax hold promise for treatment of multiple myeloma but rational combinations are needed to broaden efficacy. This study presents mechanistic and clinical data to support addition of pitavastatin to venetoclax regimens in myeloma. The results open a new avenue for repurposing statins in blood cancer.


Subject(s)
Antineoplastic Agents , Hematologic Neoplasms , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Multiple Myeloma , Humans , Myeloid Cell Leukemia Sequence 1 Protein , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-bcl-2/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Retrospective Studies , Prospective Studies , Antineoplastic Agents/pharmacology , Hematologic Neoplasms/drug therapy
8.
J Clin Oncol ; 40(31): 3576-3586, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36029527

ABSTRACT

PURPOSE: ABBV-383, a B-cell maturation antigen × CD3 T-cell engaging bispecific antibody, has demonstrated promising results in an ongoing first-in-human phase I study (ClinicalTrials.gov identifier: NCT03933735) in patients with relapsed/refractory multiple myeloma (RRMM). Herein, we report safety and efficacy outcomes of this phase I dose escalation/expansion study. METHODS: Patients with RRMM (≥ three prior lines including a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 monoclonal antibody) were eligible. ABBV-383 was administered intravenously over 1-2 hours once every 3 weeks, without any step dosing. A 3 + 3 design with backfilling for dose escalation was used (intrapatient escalation to highest safe dose permitted) followed by initiation of dose expansion. RESULTS: As of January 8, 2022, 124 patients (dose escalation [0.025-120 mg], n = 73; dose expansion [60 mg], n = 51) have received ABBV-383; median age was 68 years (range, 35-92 years). The most common hematologic treatment-emergent adverse events (TEAEs) were neutropenia (all grades: 37%) and anemia (29%). The most common nonhematologic TEAEs were cytokine release syndrome (57%) and fatigue (30%). Seven deaths from TEAEs were reported with all considered unrelated to study drug by the investigator. For all efficacy-evaluable patients (n = 122; all doses), the objective response rate (ORR) was 57% and very good partial response (VGPR) or better (≥ VGPR) rate was 43%. In the 60 mg dose expansion cohort (n = 49), the ORR and ≥ VGPR rates were 59% and 39%, respectively; and in the ≥ 40 mg dose escalation plus dose expansion cohorts (n = 79) were 68% and 54%, respectively. CONCLUSION: ABBV-383 in patients with RRMM was well tolerated with an ORR of 68% at doses ≥ 40 mg. This novel therapy's promising preliminary antitumor activity in heavily pretreated patients warrants further clinical evaluation.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Humans , Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , B-Cell Maturation Antigen , Neoplasm Recurrence, Local/drug therapy , Antineoplastic Agents/therapeutic use , T-Lymphocytes
9.
Clin Lymphoma Myeloma Leuk ; 21(11): 775-784, 2021 11.
Article in English | MEDLINE | ID: mdl-34551886

ABSTRACT

BACKGROUND: Venetoclax is a selective BCL-2 inhibitor with clinical activity in relapsed/refractory multiple myeloma (RRMM). Combinations of venetoclax with agents that have complementary mechanisms of action may improve venetoclax efficacy in RRMM. This study evaluated venetoclax with pomalidomide and dexamethasone (VenPd) in RRMM. PATIENTS AND METHODS: This phase II open label study (NCT03567616) evaluated VenPd in patients with RRMM who had received ≥ 1 prior therapy and were refractory to lenalidomide. Venetoclax was administered orally daily for days 1 to 28, pomalidomide was administered orally daily for days 1 to 21, and dexamethasone was administered weekly for each 28-day cycle. The primary objective was to characterize the safety and tolerability of VenPd. The secondary objectives were to evaluate the efficacy and pharmacokinetics. The study was terminated early due to partial clinical hold and decision to pursue biomarker driven strategy. RESULTS: Eight patients were enrolled. Patients had a median age of 67.5 years. All patients received 400 mg venetoclax; 4 patients experienced dose-limiting toxicities and the dose was not escalated. All patients had a grade ≥ 3 adverse event, and the most common was neutropenia (n = 6); cytopenias were the most prevalent adverse events. Five patients (63%) had a confirmed response, and the median duration of response was 12.9 months. The median progression-free survival was 10.5 months. CONCLUSIONS: Given the limited enrollment, no clear safety or efficacy conclusions about VenPd can be drawn. Preliminary safety data, particularly the occurrence of cytopenias, can be used to guide dosing strategies for future combinations of venetoclax with immunomodulatory agents.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Dexamethasone/therapeutic use , Multiple Myeloma/drug therapy , Sulfonamides/therapeutic use , Thalidomide/analogs & derivatives , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Dexamethasone/pharmacology , Female , Humans , Male , Multiple Myeloma/mortality , Progression-Free Survival , Sulfonamides/pharmacology , Thalidomide/pharmacology , Thalidomide/therapeutic use
10.
Blood Adv ; 5(19): 3748-3759, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34470049

ABSTRACT

Proteins in the antiapoptotic B-cell lymphoma 2 (BCL-2) family play a role in the pathophysiology of multiple myeloma (MM). Venetoclax is a highly selective, potent, oral BCL-2 inhibitor that induces apoptosis of MM cells, and its efficacy may be potentiated through combination with agents that increase BCL-2 dependency or have complementary mechanisms of action. The safety, tolerability, pharmacokinetics, and antitumor activity of venetoclax in combination with carfilzomib and dexamethasone (VenKd) in adults with relapsed/refractory MM (RRMM) were investigated in this phase 2 dose-escalation study. Oral venetoclax (400 or 800 mg) was administered daily in combination with intravenous carfilzomib (27, 56, or 70 mg/m2) and oral dexamethasone (20 or 40 mg) in 4 dose-finding cohorts. The expansion cohort received venetoclax 800 mg, carfilzomib 70 mg/m2, and dexamethasone 40 mg. Forty-nine patients received treatment. Median prior lines of therapy was 1 (range, 1-3), and median time in the study was 27 months. The most common treatment-emergent adverse events were diarrhea (65%), fatigue (47%), nausea (47%), and lymphopenia (35%). Serious adverse events occurred in 26 (53%) patients. Of 3 treatment-emergent deaths, 1 was considered treatment related. The overall response rate was 80% in all patients, 92% in patients with t(11;14) (n = 13), and 75% in patients without (n = 36). The rate of complete response or better was 41%. Median progression-free survival was 22.8 months. Treatment with VenKd was well tolerated and showed promising response rates in this RRMM patient population, with greater responses observed in patients with t(11;14). This trial is registered at www.clinicaltrials.gov as #NCT02899052.


Subject(s)
Multiple Myeloma , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bridged Bicyclo Compounds, Heterocyclic , Dexamethasone/therapeutic use , Humans , Multiple Myeloma/drug therapy , Oligopeptides , Sulfonamides
11.
Mol Cancer Ther ; 20(6): 999-1008, 2021 06.
Article in English | MEDLINE | ID: mdl-33785651

ABSTRACT

Since gaining approval for the treatment of chronic lymphocytic leukemia (CLL), the BCL-2 inhibitor venetoclax has transformed the treatment of this and other blood-related cancers. Reflecting the large and hydrophobic BH3-binding groove within BCL-2, venetoclax has significantly higher molecular weight and lipophilicity than most orally administered drugs, along with negligible water solubility. Although a technology-enabled formulation successfully achieves oral absorption in humans, venetoclax tablets have limited drug loading and therefore can present a substantial pill burden for patients in high-dose indications. We therefore generated a phosphate prodrug (3, ABBV-167) that confers significantly increased water solubility to venetoclax and, upon oral administration to healthy volunteers either as a solution or high drug-load immediate release tablet, extensively converts to the parent drug. Additionally, ABBV-167 demonstrated a lower food effect with respect to venetoclax tablets. These data indicate that beyond-rule-of-5 molecules can be successfully delivered to humans via a solubility-enhancing prodrug moiety to afford robust exposures of the parent drug following oral dosing.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Prodrugs/therapeutic use , Sulfonamides/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cross-Over Studies , Female , Healthy Volunteers , Humans , Prodrugs/pharmacology , Sulfonamides/pharmacology
12.
J Cell Biol ; 156(5): 905-19, 2002 Mar 04.
Article in English | MEDLINE | ID: mdl-11864993

ABSTRACT

Members of the protein kinase C (PKC) isozyme family are important signal transducers in virtually every mammalian cell type. Within the heart, PKC isozymes are thought to participate in a signaling network that programs developmental and pathological cardiomyocyte hypertrophic growth. To investigate the function of PKC signaling in regulating cardiomyocyte growth, adenoviral-mediated gene transfer of wild-type and dominant negative mutants of PKC alpha, beta II, delta, and epsilon (only wild-type zeta) was performed in cultured neonatal rat cardiomyocytes. Overexpression of wild-type PKC alpha, beta II, delta, and epsilon revealed distinct subcellular localizations upon activation suggesting unique functions of each isozyme in cardiomyocytes. Indeed, overexpression of wild-type PKC alpha, but not betaI I, delta, epsilon, or zeta induced hypertrophic growth of cardiomyocytes characterized by increased cell surface area, increased [(3)H]-leucine incorporation, and increased expression of the hypertrophic marker gene atrial natriuretic factor. In contrast, expression of dominant negative PKC alpha, beta II, delta, and epsilon revealed a necessary role for PKC alpha as a mediator of agonist-induced cardiomyocyte hypertrophy, whereas dominant negative PKC epsilon reduced cellular viability. A mechanism whereby PKC alpha might regulate hypertrophy was suggested by the observations that wild-type PKC alpha induced extracellular signal-regulated kinase1/2 (ERK1/2), that dominant negative PKC alpha inhibited PMA-induced ERK1/2 activation, and that dominant negative MEK1 (up-stream of ERK1/2) inhibited wild-type PKC alpha-induced hypertrophic growth. These results implicate PKC alpha as a necessary mediator of cardiomyocyte hypertrophic growth, in part, through a ERK1/2-dependent signaling pathway.


Subject(s)
Cardiomyopathy, Hypertrophic/enzymology , Cell Differentiation/physiology , Heart/growth & development , Isoenzymes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myocardium/enzymology , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Atrial Natriuretic Factor/drug effects , Atrial Natriuretic Factor/metabolism , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/physiopathology , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytosol/drug effects , Cytosol/metabolism , Heart/drug effects , Heart/physiology , Immunohistochemistry , Isoenzymes/drug effects , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Myocardium/cytology , Protein Kinase C/drug effects , Protein Kinase C-alpha , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tubulin/metabolism
13.
Hum Psychopharmacol ; 24(6): 453-64, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19637397

ABSTRACT

OBJECTIVE: The acute nootropic potential of donepezil in young healthy volunteers has not been adequately investigated mainly because in previous studies: (1) effects were assessed before peak-plasma concentration (Tmax) was reached; (2) only a few cognitive processes were assessed. Here we investigated a myriad of cognitive effects of augmentation of acetylcholine using an acute dose of donepezil in healthy adults at theoretical Tmax. METHODS: This was a double-blind, placebo controlled, parallel group design study of cognitive effects of acute oral donepezil (5 mg). Subjects were tested twice after donepezil ingestion: 90 min (time that coincides with previous testing in the literature) and 210 min. (theoretical Tmax). The test battery included tasks that tap cognitive domains that are sensitive to acetylcholine manipulations. RESULTS: At both testing times donepezil improved long-term recall of prose, objects recall, recall of spatial locations, and integration of objects with their locations, some effects having been related to self-reported mood enhancement. However, improvement of performance in the central executive measure (backward digit span) occurred only at Tmax. CONCLUSION: Positive cognitive effects of acute donepezil can be observed in various cognitive domains including mood, but its full nootropic potential is more clearly found close to theoretical peak-plasma concentration.


Subject(s)
Cognition/drug effects , Indans/pharmacology , Memory/drug effects , Nootropic Agents/pharmacology , Piperidines/pharmacology , Adult , Affect/drug effects , Cholinesterase Inhibitors/pharmacokinetics , Cholinesterase Inhibitors/pharmacology , Donepezil , Double-Blind Method , Humans , Indans/pharmacokinetics , Male , Nootropic Agents/pharmacokinetics , Piperidines/pharmacokinetics , Time Factors , Young Adult
14.
Clin Pharmacokinet ; 58(8): 1091-1100, 2019 08.
Article in English | MEDLINE | ID: mdl-30949874

ABSTRACT

INTRODUCTION: Venetoclax is a selective B cell lymphoma-2 inhibitor. It is approved for treatment of chronic lymphocytic leukemia and is being investigated for other hematological malignancies. Venetoclax is predominantly eliminated by the liver; therefore, there is a need to investigate the effect of hepatic insufficiency on venetoclax pharmacokinetics. METHODS: A phase I study was carried out in 24 women with normal hepatic function or mild, moderate, or severe hepatic impairment (based on Child-Pugh scores), who received a single 50 mg dose of venetoclax with a low-fat meal. Blood samples were collected up to 120 h after venetoclax administration. Pharmacokinetic parameters were estimated using non-compartmental methods. RESULTS: Venetoclax maximum observed plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) in subjects with mild or moderate hepatic impairment were similar to subjects with normal hepatic function. Mean venetoclax AUC in subjects with severe hepatic impairment was 2.3- to 2.7-fold higher than in subjects with normal hepatic function. The half-life of venetoclax in subjects with severe hepatic impairment was approximately two-fold longer than in subjects with normal hepatic function and subjects with mild or moderate hepatic impairment. Unbound fractions of venetoclax in subjects with mild, moderate, and severe hepatic impairment were similar to the subjects with normal hepatic function. No significant adverse safety events were reported. CONCLUSIONS: No venetoclax dosage adjustment is required in subjects with mild or moderate hepatic impairment. In subjects with severe hepatic impairment, a 50% dose reduction of venetoclax is recommended to account for higher exposures and the longer half-life.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Hepatic Insufficiency/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacokinetics , Administration, Oral , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/blood , Case-Control Studies , Female , Half-Life , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Middle Aged , Safety , Sulfonamides/administration & dosage , Sulfonamides/blood
15.
Mol Cell Biol ; 25(3): 865-78, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15657416

ABSTRACT

MEK1, a member of the mitogen-activated protein kinase (MAPK) cascade that directly activates extracellular signal-regulated kinase (ERK), induces cardiac hypertrophy in transgenic mice. Calcineurin is a calcium-regulated protein phosphatase that also functions as a positive regulator of cardiac hypertrophic growth through a direct mechanism involving activation of nuclear factor of activated T-cell (NFAT) transcription factors. Here we determined that calcineurin-NFAT and MEK1-ERK1/2 signaling pathways are interdependent in cardiomyocytes, where they directly coregulate the hypertrophic growth response. For example, genetic deletion of the calcineurin Abeta gene reduced the hypertrophic response elicited by an activated MEK1 transgene in the heart, while inhibition of calcineurin or NFAT in cultured neonatal cardiomyocytes also blunted the hypertrophic response driven by activated MEK1. Conversely, targeted inhibition of MEK1-ERK1/2 signaling in cultured cardiomyocytes attenuated the hypertrophic growth response directed by activated calcineurin. However, targeted inhibition of MEK1-ERK1/2 signaling did not directly affect calcineurin-NFAT activation, nor was MEK1-ERK1/2 activation altered by targeted inhibition of calcineurin-NFAT. Mechanistically, we show that MEK1-ERK1/2 signaling augments NFAT transcriptional activity independent of calcineurin, independent of changes in NFAT nuclear localization, and independent of alterations in NFAT transactivation potential. In contrast, MEK1-ERK1/2 signaling enhances NFAT-dependent gene expression through an indirect mechanism involving induction of cardiac AP-1 activity, which functions as a necessary NFAT-interacting partner. As a second mechanism, MEK1-ERK1/2 and calcineurin-NFAT proteins form a complex in cardiac myocytes, resulting in direct phosphorylation of NFATc3 within its C terminus. MEK1-ERK1/2-mediated phosphorylation of NFATc3 directly augmented its DNA binding activity, while inhibition of MEK1-ERK1/2 signaling reduced NFATc3 DNA binding activity. Collectively, these results indicate that calcineurin-NFAT and MEK1-ERK1/2 pathways constitute a codependent signaling module in cardiomyocytes that coordinately regulates the growth response through two distinct mechanisms.


Subject(s)
Calcineurin/metabolism , Cell Enlargement , DNA-Binding Proteins/metabolism , MAP Kinase Kinase 1/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Cells, Cultured , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/pathology , NFATC Transcription Factors , Phosphorylation , Protein Binding , Rats , Signal Transduction/physiology , Transcriptional Activation
16.
Arq Neuropsiquiatr ; 66(2B): 312-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18641862

ABSTRACT

INTRODUCTION: Semantic relations among words and repetition enhance free recall, but it is unknown if these facilitating factors are effective in dementia. METHOD: Alzheimer's patients (MILD-Alz, MOD-Alz) were compared to healthy elderly. Fifteen-word lists were read out to the subjects. In four sets of lists the words in intermediary input positions were semantically related or not, or the midlist words were repeated, or they were repeated and semantically related. RESULTS: The usual third peak of recall of semantically related words was not observed in MOD-Alz, repetition of words did not increase recall of the patients, and the combination of relatedness and repetition benefited only MID-Alz. In a second experiment, with related or unrelated midlist words, and list length shortened from 15 to 9 words, semantic facilitation was observed in mild and moderate Alzheimer s patients, although diminished compared to controls. CONCLUSION: Progression of dementia turns facilitating factors of recall less effective.


Subject(s)
Alzheimer Disease/psychology , Language Disorders/psychology , Mental Recall/physiology , Semantics , Verbal Learning/physiology , Aged , Alzheimer Disease/physiopathology , Analysis of Variance , Case-Control Studies , Educational Status , Humans , Language Disorders/physiopathology , Neuropsychological Tests , Paired-Associate Learning/physiology , Serial Learning/physiology
17.
Adv Ther ; 35(11): 2015-2023, 2018 11.
Article in English | MEDLINE | ID: mdl-30264382

ABSTRACT

INTRODUCTION: Venetoclax, a substrate of cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp), is approved for the treatment of patients with chronic lymphocytic leukemia who have received at least one prior therapy. This study evaluated the effect of azithromycin, a commonly used antibiotic in cancer patients and a P-gp inhibitor, on the pharmacokinetics of venetoclax. METHODS: In this single-center, open-label, nonfasting, two-period study, 12 healthy female subjects received a single 100 mg dose of venetoclax on day 1 of period 1 and day 3 of period 2. Subjects received azithromycin 500 mg on day 1 and 250 mg once daily on days 2 through 5. Serial blood samples for the determination of venetoclax concentrations were collected after dosing in both periods. Safety was evaluated throughout the study. RESULTS: Following coadministration of venetoclax with multiple doses of azithromycin, venetoclax maximum concentration and area under the curve to infinite time were 25% and 35% lower, respectively, compared to venetoclax administered alone. Venetoclax half-life and time to maximum concentration remained relatively unchanged when administered with azithromycin. Venetoclax was well tolerated with no serious adverse events reported. CONCLUSIONS: The modest changes in venetoclax exposures when given with azithromycin indicate that no dose adjustment would be needed when venetoclax is coadministered with azithromycin or other drugs with P-gp inhibitory potential. Azithromycin represents an alternative to other antimicrobial agents with higher potential to alter venetoclax pharmacokinetics such as clarithromycin, erythromycin, and ciprofloxacin. FUNDING: AbbVie in collaboration with Genentech/Roche.


Subject(s)
Azithromycin/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Sulfonamides/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Adult , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Azithromycin/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cytochrome P-450 CYP3A/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Drug Monitoring/methods , Female , Healthy Volunteers , Humans , Sulfonamides/administration & dosage
18.
Clin Pharmacol Drug Dev ; 7(4): 435-440, 2018 05.
Article in English | MEDLINE | ID: mdl-29058801

ABSTRACT

Venetoclax has been approved in the United States, Europe, Canada, and Australia for appropriate patients with difficult-to-treat chronic lymphocytic leukemia (CLL). The objective of this phase 1 study was to evaluate the pharmacokinetics of venetoclax in Chinese subjects to inform the dose selection of venetoclax in a phase 2 study of patients with relapsed/refractory (R/R) CLL in China. Twelve healthy first-generation Han Chinese subjects received a single 100-mg dose of venetoclax following a low-fat breakfast. Pharmacokinetic parameters were estimated using noncompartmental methods. After a single dose of venetoclax in healthy Chinese subjects, the median time to peak concentration was 6 hours (range, 4 to 6 hours), and the mean ± SD Cmax , AUCinf , and terminal half-life were 1.0 ± 0.32 µg/mL, 12.6 ± 5.4 µg·h/mL, and 18.4 ± 2.97 hours, respectively. On average, venetoclax Cmax and AUCinf values were 94% and 66% higher, respectively, in Chinese subjects compared with those observed historically for non-Asian subjects receiving the same dose. Based on these pharmacokinetic results and the established exposure-response relationship of venetoclax in non-Asian CLL subjects, a 400-mg once-daily dosage regimen was selected for evaluating the venetoclax pharmacokinetics, efficacy, and safety in the venetoclax phase 2 open-label study in Chinese subjects with R/R CLL.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacokinetics , Adult , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , China/ethnology , Drug Dosage Calculations , Female , Half-Life , Healthy Volunteers , Humans , Male , Middle Aged , Sulfonamides/administration & dosage
19.
J Clin Invest ; 111(10): 1475-86, 2003 May.
Article in English | MEDLINE | ID: mdl-12750397

ABSTRACT

The MAPKs are important transducers of growth and stress stimuli in virtually all eukaryotic cell types. In the mammalian heart, MAPK signaling pathways have been hypothesized to regulate myocyte growth in response to developmental signals or physiologic and pathologic stimuli. Here we generated cardiac-specific transgenic mice expressing dominant-negative mutants of p38alpha, MKK3, or MKK6. Remarkably, attenuation of cardiac p38 activity produced a progressive growth response and myopathy in the heart that correlated with the degree of enzymatic inhibition. Moreover, dominant-negative p38alpha, MKK3, and MKK6 transgenic mice each showed enhanced cardiac hypertrophy following aortic banding, Ang II infusion, isoproterenol infusion, or phenylephrine infusion for 14 days. A mechanism underlying this enhanced-growth profile was suggested by the observation that dominant-negative p38alpha directly augmented nuclear factor of activated T cells (NFAT) transcriptional activity and its nuclear translocation. In vivo, NFAT-dependent luciferase reporter transgenic mice showed enhanced activation in the presence of the dominant-negative p38alpha transgene before and after the onset of cardiac hypertrophy. More significantly, genetic disruption of the calcineurin Abeta gene rescued hypertrophic cardiomyopathy and depressed functional capacity observed in p38-inhibited mice. Collectively, these observations indicate that reduced p38 signaling in the heart promotes myocyte growth through a mechanism involving enhanced calcineurin-NFAT signaling.


Subject(s)
Calcineurin/metabolism , Cardiomyopathy, Hypertrophic/physiopathology , DNA-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nuclear Proteins , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Calcineurin/deficiency , Calcineurin/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Cells, Cultured , Disease Progression , Gene Targeting , Genes, Dominant , Genes, Reporter , In Vitro Techniques , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 14 , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NFATC Transcription Factors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Rats , Transgenes , Up-Regulation
20.
Mol Cell Biol ; 23(12): 4331-43, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12773574

ABSTRACT

Calcineurin is a calcium-regulated serine-threonine protein phosphatase that controls developmental and inducible biological responses in diverse cell types, in part through activation of the transcription factor nuclear factor of activated T cells (NFAT). In skeletal muscle, calcineurin has been implicated in the regulation of myoblast differentiation, hypertrophy of mature myofibers, and fiber type switching in response to alterations in intracellular calcium concentration. However, considerable disagreement persists about the functional role of calcineurin signaling in each of these processes. Here we evaluated the molecular phenotypes of skeletal muscle from both calcineurin Aalpha and calcineurin Abeta gene-targeted mice. Calcineurin Aalpha was observed to be the predominant catalytic isoform expressed in nearly all skeletal muscles examined. Neither calcineurin Aalpha or Abeta null mice showed any gross growth-related alterations in skeletal muscle, nor was fiber size or number altered in glycolytic/fast muscle types. In contrast, both calcineurin Aalpha and Abeta gene-targeted mice demonstrated an alteration in myofiber number in the soleus, an oxidative/slow-type muscle. More significantly, calcineurin Aalpha and Abeta gene-targeted mice showed a dramatic down-regulation in the oxidative/slow fiber type program in multiple muscles (both slow and fast). Associated with this observation, NFAT-luciferase reporter transgenic mice showed significantly greater activity in slow fiber-containing muscles than in fast. However, only calcineurin Aalpha null mice showed a defect in NFAT nuclear occupancy or NFAT-luciferase transgene activity in vivo. Collectively, our results suggest that calcineurin signaling plays a critical role in regulating skeletal muscle fiber type switching but not hypertrophy. Our results also suggest that fiber type switching occurs through an NFAT-independent mechanism.


Subject(s)
Calcineurin/genetics , Calcineurin/metabolism , Muscle, Skeletal/metabolism , Animals , Blotting, Western , Brain/metabolism , Calcineurin/chemistry , Catalytic Domain , Cell Nucleus/metabolism , Dipeptides , Down-Regulation , Hypertrophy , Immunohistochemistry , Mice , Mice, Transgenic , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/pathology , NADH Tetrazolium Reductase/pharmacology , Oxygen/metabolism , Phenotype , Protein Binding , Protein Isoforms , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL