Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Muscle Nerve ; 57(6): 921-926, 2018 06.
Article in English | MEDLINE | ID: mdl-29486514

ABSTRACT

INTRODUCTION: ACE-083 is a locally acting follistatin-based therapeutic that binds myostatin and other muscle regulators and has been shown to increase muscle mass and force in neuromuscular disease mouse models. This first-in-human study examined these effects. METHODS: In this phase 1, randomized, double-blind, placebo-controlled, dose-ranging study in healthy postmenopausal women, ACE-083 (50-200 mg) or placebo was administered unilaterally into rectus femoris (RF) or tibialis anterior (TA) muscles as 1 or 2 doses 3 weeks apart. RESULTS: Fifty-eight postmenopausal women were enrolled, 42 ACE-083 and 16 placebo. No serious adverse events (AE), dose-limiting toxicities, or discontinuations resulting from AEs occurred. Maximum (mean ± SD) increases in RF and TA muscle volume were 14.5% ± 4.5% and 8.9% ± 4.7%, respectively. No significant changes in mean muscle strength were observed. DISCUSSION: ACE-083 was well tolerated and resulted in significant targeted muscle growth. ACE-083 may have the potential to increase muscle mass in a wide range of neuromuscular disorders. Muscle Nerve 57: 921-926, 2018.


Subject(s)
Follistatin/pharmacology , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Recombinant Proteins/pharmacology , Aged , Double-Blind Method , Female , Healthy Volunteers , Humans , Middle Aged , Organ Size/drug effects
2.
Clin Pharmacol Drug Dev ; 6(4): 343-349, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27739232

ABSTRACT

Ketorolac tromethamine is a nonsteroidal anti-inflammatory drug that exhibits analgesic activity with no sedative or anxiolytic properties. Twelve healthy male subjects were enrolled in a study to receive either of 2 treatments over 2 periods in an open-label, randomized, 2-way crossover design: (A) 120 mg of ketorolac tromethamine administered as a continuous subcutaneous infusion over a 24-hour period; or (B) an identical total daily dose administered as 4 intramuscular bolus injections of 30 mg each given every 6 hours (current labeled treatment regimen). The pharmacokinetic and safety profiles were evaluated for both treatments. Both modes of administration have similar values for area under the curve (AUC) and half-life (t1/2 ), suggesting that continuous subcutaneous infusion and repeated intramuscular bolus injections have similar bioavailability. The peak plasma concentration (Cmax ) was 40% lower when ketorolac was administered as a continuous subcutaneous infusion compared with repeat intramuscular bolus injections. The concentration at steady-state (Css ) for continuous subcutaneous infusion was between the Cmax and Ctrough values obtained following the 4 intramuscular injections. Both treatment arms were well tolerated.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Ketorolac Tromethamine/administration & dosage , Ketorolac Tromethamine/pharmacokinetics , Adult , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Area Under Curve , Cross-Over Studies , Healthy Volunteers , Humans , Infusions, Subcutaneous/adverse effects , Injections, Intramuscular/adverse effects , Ketorolac Tromethamine/adverse effects , Male , Middle Aged , Young Adult
3.
Biopharm Drug Dispos ; 37(9): 542-549, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27704562

ABSTRACT

Raltegravir is a human immunodeficiency virus (HIV)-1 integrase strand transfer inhibitor currently marketed at a dose of 400 mg twice daily (BID). Raltegravir for once daily regimen (QD) at a dose of 1200 mg (2 x 600 mg) is under development and offers a new treatment option for HIV-1 infected treatment-naive subjects. Since raltegravir is eliminated mainly by metabolism via an UDP-glucuronosyltransferase (UGT) 1 A1-mediated glucuronidation pathway, co-administration of UGT1A1 inducers may alter plasma levels of raltegravir. Efavirenz, an UGT1A1 inducer, was used to assess the impact of altered UGT activity on a 1200 mg QD dose of raltegravir. An open label, randomized, 2-period fixed-sequence Phase 1 study was performed in adult healthy male and female subjects (non-childbearing potential) ≥ 19 and ≤55 years of age, with a body mass index (BMI) ≥ 18.5 and ≤32.0 kg/m2 . Subjects (n = 21) received a single oral dose of 1200 mg raltegravir at bedtime on an empty stomach on Day 1 in Period 1. After a washout period of at least 7 days, subjects received oral doses of 600 mg efavirenz QD at bedtime for 14 consecutive days in Period 2. Subjects received a single oral dose of 1200 mg raltegravir co-administered with 600 mg efavirenz on Day 12 of Period 2. Pharmacokinetic (PK) samples were collected for 72 hours following raltegravir dosing and analyzed using a validated bioanalytical method to quantify raltegravir plasma concentrations. PK parameters were estimated using non-compartmental analysis. Administration of single 1200 mg oral doses of raltegravir alone and co-administered with multiple oral doses of efavirenz were generally well tolerated in healthy subjects. Co-administration with efavirenz yielded geometric mean ratios (GMRs) and their associated 90% confidence intervals (90% CIs) for raltegravir AUC0-∞, Cmax , and C24 of 0.86 (0.73, 1.01), 0.91 (0.70, 1.17), and 0.94 (0.76, 1.17), respectively. The results show that efavirenz modestly reduced the exposure of raltegravir. The reduction in raltegravir exposure is not considered clinically meaningful. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Anti-HIV Agents/blood , Benzoxazines/blood , Glucuronosyltransferase/blood , Raltegravir Potassium/blood , Administration, Oral , Adult , Alkynes , Anti-HIV Agents/administration & dosage , Benzoxazines/administration & dosage , Cyclopropanes , Drug Interactions/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Female , Humans , Male , Middle Aged , Raltegravir Potassium/administration & dosage , Young Adult
4.
Biopharm Drug Dispos ; 37(9): 533-541, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27696440

ABSTRACT

Raltegravir is a human immunodeficiency virus (HIV)-1 integrase strand transfer inhibitor currently marketed at a dose of 400 mg twice-daily (b.i.d.). Raltegravir 1200 mg once-daily (q.d.) (investigational q.d. formulation of 2 × 600 mg tablets; q.d. RAL) was found to be generally well tolerated and non-inferior to the marketed 400 mg b.i.d. dose at 48 weeks in a phase 3 trial. Since raltegravir is eliminated mainly by metabolism via a uridine diphosphate glucuronosyltransferase (UGT) 1A1-mediated glucuronidation pathway, co-administration of UGT1A1 inhibitors may increase the plasma levels of q.d. RAL. To assess this potential, the drug interaction of 1200 mg raltegravir using atazanavir, a known UGT1A1 inhibitor, was studied. An open-label, randomized, 2-period, fixed-sequence phase 1 study was performed in adult healthy male and female (non-childbearing potential) subjects ≥ 19 and ≤ 55 years of age, with a body mass index (BMI) ≥ 18.5 and ≤ 32.0 kg/m2 . Subjects (n = 14) received a single oral dose of 1200 mg raltegravir in period 1. After a washout period of at least 7 days, the subjects received oral doses of 400 mg atazanavir q.d. for 9 consecutive days, with a single oral dose of 1200 mg raltegravir co-administered on day 7 of period 2. Serial blood samples were collected for 72 h following raltegravir dosing and analysed using a validated bioanalytical method to quantify raltegravir plasma concentrations. Co-administration with atazanavir yielded GMRs (90% CIs) for raltegravir AUC0-∞ , Cmax and C24 of 1.67 (1.34, 2.10), 1.16 (1.01, 1.33) and 1.26 (1.08, 1.46), respectively. There was no effect of raltegravir on serum total bilirubin. In contrast, atazanavir increased the mean bilirubin by up to 200%, an effect that was preserved in the atazanavir/raltegravir treatment group. Administration of single q.d. RAL alone and co-administered with multiple oral doses of atazanavir were generally well tolerated in healthy subjects. The results show that atazanavir increased the PK exposure of raltegravir; therefore, co-administration of atazanavir with raltegravir q.d. is not recommended. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Atazanavir Sulfate/blood , HIV Integrase Inhibitors/blood , HIV Protease Inhibitors/blood , Raltegravir Potassium/blood , Administration, Oral , Adult , Atazanavir Sulfate/administration & dosage , Drug Interactions/physiology , Female , HIV Integrase Inhibitors/administration & dosage , HIV Protease Inhibitors/administration & dosage , Humans , Male , Middle Aged , Raltegravir Potassium/administration & dosage , Young Adult
5.
Genes Chromosomes Cancer ; 54(12): 762-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26355392

ABSTRACT

In eosinophilia-associated myeloproliferative neoplasms (MPN-eo), constitutive activation of protein tyrosine kinases (TK) as consequence of translocations, inversions, or insertions and creation of TK fusion genes is recurrently observed. The most commonly involved TK and their potential TK inhibitors include PDGFRA at 4q12 or PDGFRB at 5q33 (imatinib), FGFR1 at 8p11 (ponatinib), and JAK2 at 9p24 (ruxolitinib). We here report the identification of three new PDGFRB fusion genes in three male MPN-eo patients: MPRIP-PDGFRB in a case with t(5;17)(q33;p11), CPSF6-PDGFRB in a case with t(5;12)(q33;q15), and GOLGB1-PDGFRB in a case with t(3;5)(q13;q33). The fusion proteins identified by 5'-rapid amplification of cDNA ends polymerase chain reaction (PCR) or DNA-based long distance inverse PCR are predicted to contain the TK domain of PDGFRB. The partner genes contain domains like coiled-coil structures, which are likely to cause dimerization and activation of the TK. In all patients, imatinib induced rapid and durable complete remissions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Eosinophilia/genetics , Gene Fusion , Membrane Proteins/genetics , Myeloproliferative Disorders/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Translocation, Genetic , mRNA Cleavage and Polyadenylation Factors/genetics , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 5/genetics , Cytogenetic Analysis , Eosinophilia/drug therapy , Eosinophilia/pathology , Golgi Matrix Proteins , Humans , Imatinib Mesylate/therapeutic use , In Situ Hybridization, Fluorescence , Male , Middle Aged , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , Polymerase Chain Reaction , Remission Induction
6.
Environ Toxicol ; 29(10): 1201-16, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23436777

ABSTRACT

Nonpersistent pesticides are considered less harmful for the environment, but their impact as endocrine disruptors has not been fully explored. The pesticide Switch was applied to grape vines, and the maximum residue concentration of its active ingredients was quantified. The transactivation potential of the pesticides Acorit, Frupica, Steward, Reldan, Switch, Cantus, Teldor, and Scala and their active compounds (hexythiazox, mepanipyrim, indoxacarb, chlorpyrifos-methyl, cyprodinil, fludioxonil, boscalid, fenhexamid, and pyrimethanil) were tested on human estrogen receptor α (ERα), androgen receptor (AR) and arylhydrocarbon receptor (AhR) in vitro. Relative binding affinities of the pure pesticide constituents for AR and their effect on human breast cancer and prostate cancer cell lines were evaluated. Residue concentrations of Switch's ingredients were below maximum residue limits. Fludioxonil and fenhexamid were ERα agonists (EC50 -values of 3.7 and 9.0 µM, respectively) and had time-dependent effects on endogenous ERα-target gene expression (cyclin D1, progesterone receptor, and nuclear respiratory factor 1) in MCF-7 human breast cancer cells. Fludioxonil, mepanipyrim, cyprodinil, pyrimethanil, and chlorpyrifos-methyl were AhR-agonists (EC50 s of 0.42, 0.77, 1.4, 4.6, and 5.1 µM, respectively). Weak AR binding was shown for chlorpyrifos-methyl, cyprodinil, fenhexamid, and fludioxonil. Assuming a total uptake which does not take metabolism and clearance rates into account, our in vitro evidence suggests that pesticides could activate pathways affecting hormonal balance, even within permitted limits, thus potentially acting as endocrine disruptors.


Subject(s)
Endocrine Disruptors/toxicity , Estrogen Receptor alpha/metabolism , Pesticides/toxicity , Receptors, Androgen/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Male , Prostatic Neoplasms/metabolism
7.
Mod Pathol ; 26(1): 106-16, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22899295

ABSTRACT

Approximately 50% of prostate cancers are characterized by TMPRSS2 (transmembrane protease serine 2)-ERG (avian v-ets erythroblastosis virus E26 oncogene homolog) gene fusions resulting in an androgen-regulated overexpression of the transcription factor ERG. Some studies have suggested prognostic or predictive relevance of ERG status in prostate cancer. Such concepts could be impaired by extensive ERG heterogeneity in analyzed tumors. The aim of this study was to analyze the extent of heterogeneity for TMPRSS2-ERG fusion in prostate cancer. To enable large-scale studies on the extent of heterogeneity of biomarkers in prostate cancer, a heterogeneity tissue microarray containing samples from 10 different tumor blocks of 190 large prostate cancers selected from a consecutive series of 480 radical prostatectomies was developed. ERG expression was analyzed by immunohistochemistry. Positive ERG immunostaining was found in arrayed cancer-containing samples from 103 of the 178 analyzable patients (58%). ERG immunostaining was homogeneously positive in 29 prostate cancers (16%), whereas heterogeneous ERG positivity was seen in 74 cancers (42%). ERG heterogeneity was within one tumor focus (intrafocal heterogeneity) in 69 cases (93% of heterogeneous cases) and between different tumor foci (interfocal heterogeneity) in 5 cases (7%). Marked intrafocal heterogeneity challenges the concept of TMPRSS2-ERG fusion always representing an early step in prostate cancer development. Marked heterogeneity also compromises the concept of analyzing ERG status for treatment decisions in diagnostic needle core biopsies.


Subject(s)
Biomarkers, Tumor/analysis , Prostatic Neoplasms/metabolism , Trans-Activators/biosynthesis , Adult , Aged , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Oncogene Proteins, Fusion/analysis , Oncogene Proteins, Fusion/biosynthesis , Prostatic Neoplasms/pathology , Tissue Array Analysis , Trans-Activators/analysis , Transcriptional Regulator ERG
8.
Int J Cancer ; 125(1): 34-42, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19326439

ABSTRACT

The phloroglucinol derivative hyperforin, a major bioactive constituent of St. John's wort, is increasingly recognized as being able to regulate a variety of pathobiological processes and, thus, to possess potential therapeutic properties. In the context of cancer, hyperforin induces the apoptosis of cancer cells, inhibits angiogenesis and suppresses metastasis formation. Here, we report a new pharmacological function of hyperforin and its stabilized derivative aristoforin, namely the suppression of lymphatic endothelial cell (LEC) growth and lymphangiogenesis. At concentrations less than 10 microM, we found that these compounds induce cell cycle arrest of LECs, and at higher concentrations induce apoptosis. The loss of mitochondrial membrane potential and the activation of caspase-9 during the induction of apoptosis indicate that the intrinsic pathway of apoptosis is stimulated by these compounds, similar to the situation in tumor cells. In thoracic duct ring outgrowth assays, hyperforin and aristoforin both inhibited lymphangiogenesis, as evidenced by the suppression of lymphatic capillary outgrowth. In an in vivo animal model, both compounds were able to inhibit tumor-induced lymphangiogenesis. Together these data substantiate a new role for hyperforin and its derivatives as suppressors of lymphangiogenesis, and support their further investigation as potential anticancer drugs that target tumor growth and metastasis at multiple levels.


Subject(s)
Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Lymphangiogenesis/drug effects , Neoplasms, Experimental/pathology , Phloroglucinol/analogs & derivatives , Terpenes/pharmacology , Animals , Aorta, Thoracic/drug effects , Apoptosis/drug effects , Bridged Bicyclo Compounds/pharmacology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Cycle/drug effects , Cells, Cultured , Cytochromes c/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Phloroglucinol/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Xenograft Model Antitumor Assays
9.
Org Biomol Chem ; 5(17): 2767-9, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17700843

ABSTRACT

A new staining reagent was prepared and its ability to stain several azide-containing agents on TLC plates was determined.


Subject(s)
Azides/chemistry , Coloring Agents/chemistry , Azides/chemical synthesis , Chromatography, Thin Layer , Coloring Agents/chemical synthesis , Molecular Structure
10.
Cancer Chemother Pharmacol ; 59(2): 157-64, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16703323

ABSTRACT

The inhibition of kinesin Eg5 by small molecules such as monastrol is currently evaluated as an approach to develop a novel class of antiproliferative drugs for the treatment of malignant tumours. Therefore, we studied the effects of the new monastrol analogues enastron, dimethylenastron and vasastrol VS-83 on the proliferation of human glioblastoma cells in the kinetic crystal violet assay. Compared to monastrol, the new cell cycle specific compounds showed an at least one order of magnitude higher anti proliferative activity against U-87 MG, U-118 MG, and U-373 MG glioblastoma cells. The compounds were neither inactivated by hydrolysis nor by binding to serum proteins. Moreover, we demonstrated the characteristic monoaster formation after incubation of cells with the new compounds by confocal laser scanning microscopy. We also showed that the arrangement of beta-actin and tubulin, vital components of the cyto-skeleton of mitotic and quiescent cells, were not affected by the new compounds. Due to the necessity of overcoming the blood-brain barrier in the treatment of brain tumours, we investigated if the new monastrol analogues are modulators or substrates of the p-glycoprotein (p-gp) 170 by a flow cytometric calcein-AM efflux assay. The tested compounds showed no modulating effects on the p-gp function. With respect to the treatment of primary and secondary CNS tumours, the results of our experiments suggest that the new monastrol analogues represent an interesting class of potential anticancer drugs, predicted to be less neurotoxic in comparison to classical tubulin inhibitors.


Subject(s)
Cell Proliferation/drug effects , Kinesins/antagonists & inhibitors , Pyrimidines/pharmacology , Thiones/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Acridines/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cysteine/analogs & derivatives , Cysteine/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry/methods , Fluoresceins/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Insecticides/pharmacology , Kinesins/metabolism , Molecular Structure , Paclitaxel/pharmacology , Pyrimidines/chemistry , Quinazolines/chemistry , Quinazolines/pharmacology , Rotenone/pharmacology , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tetrahydroisoquinolines/pharmacology , Thiones/chemistry , Time Factors , Tubulin/metabolism , Tubulin Modulators/pharmacology , Vinblastine/pharmacology
11.
Bioorg Med Chem ; 13(22): 6094-111, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16084101

ABSTRACT

The mitotic kinesin Eg5 (or KSP) is a crucial player in the development and function of the mitotic spindle. Inhibition of this protein leads to cell cycle arrest and apoptosis without interfering with other microtubule-dependent processes. Therefore, it is a potential target in cancer therapy. Here, we report the synthesis and biological evaluation of a small library of molecules based on the structure of the known Eg5 inhibitor HR22C16. One of these derivatives (compound trans-24) proved to be a potent and specific Eg5 inhibitor.


Subject(s)
Carbolines/chemical synthesis , Carbolines/pharmacology , Kinesins/antagonists & inhibitors , Mitosis/drug effects , Xenopus Proteins/antagonists & inhibitors , Animals , Carbolines/chemistry , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Indoles/chemistry , Inhibitory Concentration 50 , Kinesins/genetics , Molecular Structure , Neurotoxins/chemical synthesis , Neurotoxins/pharmacology , Phenols/chemistry , Xenopus Proteins/genetics
13.
Chembiochem ; 6(1): 171-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15593112

ABSTRACT

Hyperforin, a natural product of St. John's wort (Hypericum perforatum L.), has a number of pharmacological activities, including antidepressive and antibacterial properties. Furthermore, hyperforin has pronounced antitumor properties against different tumor cell lines, both in vitro and in vivo. Despite being a promising novel anticancer agent, the poor solubility and stability of hyperforin in aqueous solution limits its potential clinical application. In this study, we present the synthesis of hyperforin derivatives with improved pharmacological activity. The synthesized compounds were tested for their solubility and stability properties. They were also investigated for their antitumor properties, both in vitro and in vivo. One of these hyperforin derivatives, Aristoforin, is more soluble in aqueous solution than hyperforin and is additionally highly stable. Importantly, it retains the antitumor properties of the parental compound without inducing toxicity in experimental animals. These data strongly suggest that Aristoforin has potential as an anticancer drug.


Subject(s)
Antineoplastic Agents/pharmacology , Phloroglucinol/analogs & derivatives , Phloroglucinol/pharmacology , Terpenes/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Bridged Bicyclo Compounds/chemistry , Drug Stability , Phloroglucinol/chemical synthesis , Phloroglucinol/chemistry , Rats , Solubility , Terpenes/chemical synthesis , Terpenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL